Language selection

Search

Patent 2588908 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2588908
(54) English Title: INDUCTION OF NEUROGENESIS AND STEM CELL THERAPY IN COMBINATION WITH COPOLYMER 1
(54) French Title: INDUCTION DE NEUROGENESE ET THERAPIE PAR CELLULES SOUCHES EN COMBINAISON AVEC LE COPOLYMERE 1
Status: Granted
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 31/74 (2006.01)
  • A61K 38/16 (2006.01)
  • A61K 39/385 (2006.01)
  • A61P 25/00 (2006.01)
  • A61P 25/28 (2006.01)
  • A61K 35/14 (2006.01)
(72) Inventors :
  • EISENBACH-SCHWARTZ, MICHAL (Israel)
  • ARNON, RUTH (Israel)
  • BUTOVSKY, OLEG (Israel)
  • ZIV, YANIV (Israel)
  • KIPNIS, JONATHAN (Israel)
  • RON, NOGA (Israel)
  • EILAM, RAYA (Israel)
  • AHARONI, RINA (Israel)
(73) Owners :
  • YEDA RESEARCH AND DEVELOPMENT CO. LTD (Israel)
(71) Applicants :
  • YEDA RESEARCH AND DEVELOPMENT CO. LTD (Israel)
(74) Agent: OSLER, HOSKIN & HARCOURT LLP
(74) Associate agent:
(45) Issued: 2014-09-23
(86) PCT Filing Date: 2005-11-29
(87) Open to Public Inspection: 2006-06-01
Examination requested: 2010-11-25
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/IL2005/001275
(87) International Publication Number: WO2006/057003
(85) National Entry: 2007-05-29

(30) Application Priority Data:
Application No. Country/Territory Date
60/631,163 United States of America 2004-11-29
60/690,498 United States of America 2005-06-15

Abstracts

English Abstract




A method for inducing and enhancing neurogenesis and /or oligodendrogenesis
from endogenous as well as from exogenously administered stem cells comprises
administering to an individual in need thereof an agent selected from the
group consisting of Copolymer 1, a Copolymer 1-related polypeptide, a
Copolymer 1, a Copolymer 1-related polypeptide, or a Copolymer 1-related
peptide. The method is particularly useful for stem cell therapy in
combination with the agent.


French Abstract

L'invention concerne un procédé destiné à induire et à améliorer la neurogenèse et / ou l'oligodendrogenèse provenant de cellules souches administrées de façon endogène comme de façon exogène; le procédé consiste à administrer à un individu un agent sélectionné dans le groupe constitué d'un copolymère 1, d'un polypeptide relié au copolymère 1 ou d'un peptide relié au copolymère 1, et de lymphocytes T activés par le copolymère 1, le polypeptide relié au copolymère 1 ou le peptide relié au copolymère 1. Le procédé est particulièrement utile dans la thérapie par cellules souches en combinaison avec l'agent.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS
1. Use of Copolymer 1 or a pharmaceutically acceptable salt thereof
in combination with stem cell therapy for treatment of Parkinson's disease;
Huntington's disease; Alzheimer's disease, multiple sclerosis; or
amyotrophic lateral sclerosis (ALS).
2. The use according to claim 1, wherein said Copolymer 1 is for
administration to the patient before, concomitantly or after the
transplantation of the stem cells to said patient.
3. The use according to claim 1 or 2, wherein the stem cells are adult
stem cells, embryonic stem cells, umbilical cord blood stem cells,
hematopoietic stem cells, peripheral blood stem cells, mesenchimal stem
cells, multipotent stem cells, neural stem cells, neural progenitor cells,
stromal stem cells, progenitor cells, or genetically-engineered stem cells.
115

Description

Note: Descriptions are shown in the official language in which they were submitted.


DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPREND PLUS D'UN TOME.
CECI EST LE TOME 1 DE 2
NOTE: Pour les tomes additionels, veillez contacter le Bureau Canadien des
Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.
THIS IS VOLUME 1 OF 2
NOTE: For additional volumes please contact the Canadian Patent Office.

CA 02588908 2010-12-07
INDUCTION OF NEUROGENESIS AND STEM CELL THERAPY IN
COMBINATION WITH COPOLYMER 1
FIELD OF THE INVENTION
The present invention relates to use of a neuroprotective agent, in particular
Copolymer 1, and compositions thereof, for induction and/or enhancement of
endogenous
neurogenesis and/or oligodendrogenesis and for stem cell therapy in injuries,
diseases,
disorders or conditions, in particular those associated with the central
nervous system
(CNS) or peripheral nervous system (PNS).
Abbreviations: AP, P-amyloid; AD, Alzheimer's disease; BDNF, brain-derived
neurotrophic factor; BMS, Basso motor score; BrdU, 5-bromo-2'-deoxyuridine;
CFA,
complete Freund's adjuvant; CNS, central nervous system; Cop 1, Copolymer 1,
same as
GA; DCX, doublecortin; DG, dentate gyrus; EAE, experimental autoimmune
encephalomyelitis; EGF, epidermal growth factor; FCS, fetal calf serum; FGF,
fibroblast
growth factor; i.c.v., intracerebroventricular; GA, glatiramer acetate; GFAP,
glial
fibrillary acidic protein; GFP, green fluorescent protein; IB4, isolectin B4;
IFA,
incomplete Freund's adjuvant; IGF-I, insulin-like growth factor 1; IFN,
interferon; IL,
interleukin; LPS, lipopolysaccharide; MBP, myelin basic protein; MG,
microglia; MHC-
II, class II major histocompatibility complex molecules; MOG, myelin
oligodendrocyte
glycoprotein; MS, multiple sclerosis; MWM, Morris water maze; NeuN, neuronal
nuclear
antigen; NPC, neural stem/progenitor cell; OB, olfactory bulb; PBS, phosphate-
buffered
saline; PDL, poly-D-lysine; PNS, peripheral nervous system; RMS, rostral
migratory
stream; SCI, spinal cord injury; SGZ, subgranular zone; SVZ, subventricular
zone; TGF-
13, transforming growth factor-13; TNF, tumor necrosis factor.
1

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
BACKGROUND OF THE INVENTION
The central nervous system (CNS) is particularly vulnerable to insults that
result in cell death or damage in part because cells of the CNS have a limited

capacity for repair. Since damaged brain tissue does not regenerate, recovery
must
come from the remaining intact brain.
Poor recovery from acute insults or chronic degenerative disorders in the
CNS has been attributed to lack of neurogenesis, limited regeneration of
injured
nerves, and extreme vulnerability to degenerative conditions. The absence of
neurogenesis was explained by the assumption that soon after birth the CNS
reaches
a permanently stable state, needed to maintain the equilibrium of the brain's
complex tissue network. Research during the last decade showed, however, that
the
brain is capable of neurogenesis throughout life, albeit to a limited extent
(Morshead et al., 1994). In the inflamed brain, neurogenesis is blocked
(Ekdahl et
al., 2003; Monje et al., 2003). This latter finding strengthened the
traditional view
that local immune cells in the CNS have an adverse effect on neurogenesis.
Likewise, the limited regeneration and excessive vulnerability of CNS neurons
under inflammatory conditions or after an acute insult were put down to the
poor
ability of the CNS to tolerate the immune-derived defensive activity that is
often
associated with local inflammation and cytotoxicity mediated, for example, by
tumor necrosis factor (TNF)-a or nitric oxide (Merrill et al., 1993). More
recent
studies have shown, however, that although an uncontrolled local immune
response
indeed impairs neuronal survival and blocks repair processes, a local immune
response that is properly controlled can support survival and promote recovery

(Hauben and Schwartz, 2003; Schwartz et al., 2003). It was further shown that
after
an injury to the CNS, a local immune response that is well controlled in time,
space,
and intensity by peripheral adaptive immune processes (in which CD4+ helper T
cells are directed against self-antigens residing at the site of the lesion)
is a critical
requirement for post-traumatic neuronal survival and repair (Moalem et al.,
1999;
Butovsky et al., 2001; Schwartz et al., 2003; Shaked et al., 2004). These and
other
2

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
results led the inventor M Schwartz and colleagues to formulate the concept of

'protective autoimmunity' (Moalem et al., 1999).
Neurogenesis occurs throughout life in adult individuals, albeit to a limited
extent. Most of the newly formed cells die within the first 2-3 weeks after
proliferation and only a few survive as mature neurons. Little is known about
the
mechanism(s) underlying the existence of neural stem/progenitor cells (NPCs)
in an
adult brain and why these cells are restricted in amount and limited to
certain areas.
Moreover, very little is known about how neurogenesis from an endogenous NPC
pool can be physiologically increased. Knowledge of the factors allowing such
stem
cells to exist, proliferate, and differentiate in the adult individual is a
prerequisite for
understanding and promoting the conditions conducive to CNS repair. This in
turn
can be expected to lead to the development of interventions aimed at boosting
neural cell renewal from the endogenous stem-cell pool or from exogenously
applied stem cells.
Experiments with rat and mouse models in our laboratory have shown that
well-controlled implantation of specifically activated blood-borne macrophages

(Rapalino et al., 1998) or dendritic cells (Hauben et al., 2003) promotes
recovery
from spinal cord injury (SCI). Other studies showed that the well-controlled
activity
of autoimmune T cells reactive to CNS antigens residing in the lesioned site
can
promote recovery from axonal insults (Hauben et al., 2000; Moalem et al.,
1999). It
was also shown that neuroprotection, mediated by T cells directed specifically
to
CNS-related autoantigens, is the body's physiological response to CNS injury
(Yoles et al., 2001a, 2001b).
Under normal conditions in the adult brain, new neurons are formed in the
neurogenic niches of the subventricular zone of the lateral ventricles and the
subgranular zone of the hippocampal dentate gyrus (Kempermann et al., 2004).
Under pathological conditions some neurogenesis can also be induced in non-
neurogenic brain areas. Several studies have demonstrated, for example, that
injury
to the CNS in animals is followed by recruitment of endogenous NPCs, which can
undergo differentiation to neurons and glia at the injured site (Nakatomi et
al., 2002;
3

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Imitola et al., 2004a). However, this injury-triggered cell renewal from
endogenous
progenitors is limited in extent and is not sufficient for full replacement of
the
damaged tissue. To overcome the deficit, scientists are currently seeking ways
to
promote recovery by transplanting cultured adult NPCs (aNPCs) (McDonald et
al.,
2004). Exogenous aNPCs might contribute to recovery by acting as a source of
new
neurons and glia in the injured CNS (Cummings et al., 2005; Lepore and
Fischer,
2005) or by secreting factors that directly or indirectly promote
neuroprotection
(Lu et al., 2005) and neurogenesis from endogenous stem-cell pools (Enzmann et

al., 2005).
Current opinions concur that neurogenesis persists in the adult brain, where
it
may contribute to repair and recovery after injury. Brain insults such as
cerebral
ischemia (Jin et al., 2003), apoptosis (Magavi et al., 2000) or autoimmune
inflammatory demyelination (Picard-Riera et al., 2002) enhance neurogenesis.
Hence, multipotent cells located in the hippocampus hilus and the
subventricular
zone (SVZ) of the lateral ventricle manifest increased proliferation and
migration in
pathological situations. Moreover, progenitor cells from the SVZ that migrate
through the rostral migratory stream (RMS) to the olfactory bulb (OB) can be
triggered to differentiate into astrocytes and neurons (Picard-Riera et al.,
2004).
Nevertheless, the therapeutic significance of self-neurogenesis in CNS
pathology is
limited, as it fails to regenerate functional neurons that compensate the
damage.
In multiple sclerosis (MS) and its animal model experimental autoimmune
encephalomyelitis (EAE), the immune system provokes the detrimental process
via
autoimmune inflammatory mechanisms (Hellings et al., 2002; Behi et al., 2005).

Still, neuronal and axonal degeneration, initiated at disease onset and
revealed when
compensatory CNS resources are exhausted, are the major determinant of the
irreversible neurological disability (Bjartmar et al., 2003), particularly in
the myelin
oligodendrocyte glycoprotein (MOG) induced model (Hobom et al., 2003). Current

treatments for MS are effective in ameliorating the immune inflammatory
process,
but their ability to enhance the intrinsic CNS repair mechanism and to induce
effective neuroprotection and neurogenesis has not been shown.
4

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
A potential approach for treatment of CNS damage includes the use of adult
neural stem cells or any type of stem cells. The adult neural stem cells are
progenitor cells present in the mature mammalian brain that have the ability
of self-
renewal and, given the appropriate stimulation, can differentiate into brain
neurons,
astrocytes and oligodendrocytes. Stem cells (from other tissues) have
classically
been defined as pluripotent and having the ability to self-renew, to
proliferate, and
to differentiate into multiple different phenotype lineages. Hematopoietic
stem cells
are defined as stem cells that can give rise to cells of at least one of the
major
hematopoietic lineages in addition to producing daughter cells of equivalent
potential. Three major lineages of blood cells include the lymphoid lineage,
e.g. B-
cells and T-cells, the myeloid lineage, e.g. monocytes, granulocytes and
megakaryocytes, and the erythroid lineage, e.g. red blood cells. Certain
hematopoietic stem cells are capable of differentiating to other cell types,
including
brain cells.
Transplantation of multipotent (stern) precursor cells is a promising strategy
for the therapy of various disorders caused by loss or malfunction of single
or few
cell types. These include neurological disorders such as spinal cord injury,
subcortical neurodegenerations e.g. Huntington and Parkinson, and
demyelinatindg
diseases e.g. MS, as well as other pathological conditions such as diabetes,
myocardial infarction of cardiac failure, tissue injury and insufficient wound
healing. Particularly in MS and its animal model EAE, stem cells
differentiating
into oligodendrocytes and neurons may lead to repair of myelin damage and
replace
degenerating neurons. However, hitherto stem cell transplantation in these
systems
resulted in poor therapeutic outcome. Thus, stem cells transplanted as such
into
EAE mice were found mainly around the injection site (in cases of local
administration) or in perivascular position (when systemic administration was
employed), and their proliferation, migration and differentiation were
insufficient to
compensate for the damage inflicted by the disease (Goldman, 2005; Pluchino
and
Martini, 2005). A clinical trial in which stem cells were transplanted into
demyelinating brain areas of MS patients was discontinued in 2003, as no
evidence
5

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
of stem cell survival was found in the implanted patients (Pluchino and
Martini,
2005). These outcomes were related to the chronic inflammatory processes that
destroy the transplanted as well as the resident cells. It has been suggested
that stem
cell-based therapeutic strategies, especially those intended for MS, will
require
disease modification adjuncts to cell delivery. Stem cell therapy is also
considered
for many other medical applications, not related to neurological disorders.
Separation and cloning of neural stem cell lines from both the murine and
human brain have been reported. Human CNS neural stem cells, like their rodent

homologues, when maintained in a mitogen-containing (typically epidermal
growth
factor or epidermal growth factor plus basic fibroblast growth factor), serum-
free
culture medium, grow in suspension culture to form aggregates of cells known
as
neurospheres. Upon removal of the mitogens and provision of a substrate, the
stem
cells differentiate into neurons, astrocytes and oligodendrocytes. When such
stem
cells are reintroduced into the developing or mature brain, they can undergo
through
division, migration and growth processes, and assume neural phenotypes,
including
expression of neurotransmitters and growth factors normally elaborated by
neurons.
Thus, use of neural stem cells may be advantageous for CNS damage recovery in
at
least two ways: (1) by the stem cells partially repopulating dead areas and
reestablishing neural connections lost by CNS damage, and (2) by secretion of
important neurotransmitters and growth factors required by the brain to rewire
after
CNS damage.
Recently, a renewable source of neural stem cells was discovered in the adult
human brain. These cells may be a candidate for cell-replacement therapy for
nervous system disorders. The ability to isolate these cells from the adult
human
brain raises the possibility of performing autologous neural stem cell
transplantation. It has been reported that clinical trials with adult human
neural stern
cells have been initiated for treatment of Parkinson's disease patients. If
adult neural
stem cells are to be used in clinical trials they must be amenable to
expansion into
clinically significant quantities. Unfortunately, these cells seem to have a
limited
6

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
life-span in the culture dish and it remains to be determined whether they are
stable
at later passages and capable of generating useful numbers of neurons.
The brain has long been viewed as an immune-privileged site. However,
autoimmune T cells (controlled with respect to the onset, duration, and
intensity of
their activity) were recently shown to exert a beneficial effect on neuronal
survival
after CNS injury (Schwartz et al., 2003), as well as in cases of mental
dysfunction
(Kipnis et al., 2004). Moreover, in-depth understanding of the mechanisms
underlying the beneficial effect of T cells for degenerative neural tissue has
has
pointed out that the T cells instruct the microglia, at the injured area, to
acquire a
phenotype supportive of neural tissue. In addition, it appeared that several
immune-
based intervention can boost this protective response, all of which converts
to
microglial activation (Shaked et al., 2004). The type of damage does not
determine
the choice of the approach, it is the site which determines it. Some antigens
cross-
react with numerous antigens and thus can overcome tissue specificity barrier.
According to the present invention, we show that the same manipulation that
leads
to neuronal survival leads to neurogenesis and oligodendrogenesis. It appears
that
the same microglia, activated by T cells or by their cytokines, not only
support
neuronal survival but also support oligodendrogenesis and neurogenesis. These
results indicate that T-cell-based manipulation will create conditions in
damaged
neural tissue that favor cell renewal not only from endogenous stern cell
resources
but also from exogenously applied stem cells.
Copolymer 1 or Cop 1, a non-pathogenic synthetic random copolymer
composed of the four amino acids: L-Glu, L-Lys, L-Ala, and L-Tyr. Glatiramer
acetate (GA), one form of Cop 1, is currently an approved drug for the
treatment of
multiple sclerosis under the name of Copaxone (a trademark of Teva
Pharmaceutical Industries Ltd., Petach Tikva, Israel). It exerts a marked
suppressive
effect on EAE induced by various encephalitogens, in several species (Arnon
and
Sela, 2003).
7

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Cop 1 is a very well tolerated agent with only minor adverse reactions and
high safety profile. Treatment with Cop 1 by ingestion or inhalation is
disclosed in
US 6,214,791.
Recently it was found that in animal models Cop 1 provides a beneficial
effect for several additional disorders. Thus, Cop 1 suppresses the immune
rejection
manifested in graft-versus-host disease (GVHD) in case of bone marrow
transplantation (Schlegel et al., 1996; Aharoni et al., 1997; US 5,858,964),
as well
as in graft rejection in case of solid organ transplantation (Aharoni et al.,
2001,
2004) and in the applicant's patent applications WO 00/27417 and W0/009333A2)
WO 01/52878 and WO 01/93893 of the same applicants disclose that Cop 1,
Cop 1-related peptides and polypeptides and T cells activated therewith
protect
CNS cells from glutamate toxicity and prevent or inhibit neuronal degeneration
or
promote nerve regeneration in the CNS and in the PNS. Thus, for example, Cop 1
is ,
under evaluation as a therapeutic vaccine for neurodegenerative diseases such
as
optic neuropathies and glaucoma (Kipnis and Schwartz, 2002).
Cop 1 has been shown to act as a low-affinity antigen that activates a wide
range of self-reacting T cells, resulting in neuroprotective autoimmunity that
is
effective against both CNS white matter and grey matter degeneration (Schwartz

and Kipnis, 2002). The neuroprotective effect of Cop 1 vaccination was
demonstrated in animal models of acute and chronic neurological disorders such
as
optic nerve injury (Kipnis et al., 2000), head trauma (Kipnis et al., 2003),
glaucoma
(Schori et al., 2001; Bakalash et al., 2003), amyotrophic lateral sclerosis
(Angelov
et al., 2003) and in the applicant's patent applications WO 01/52878, WO
01/93893
and WO 03/047500.
The use of Copolymer 1 for treatment of prion-related diseases is disclosed
in WO 01/97785. Gendelman and co-workers disclose that passive immunization
with splenocytes of mice immunized with Cop 1 confers dopaminergic
neuroprotection in MPTP-treated mice (Benner et al., 2004).
8

CA 02588908 2010-12-07
Cop 1 and related copolymers and peptides have been disclosed in WO 00/05250
(Aharoni et al., 2000) for treating autoimmune diseases and in WO 2004/064717
for
treatment of Inflammatory Bowel Diseases (Aharoni et al, 2004).
The immunomodulatory effect of GA was attributed to its ability to induce
Th2/3
cells that secrete high levels of anti-inflammatory cytokines (Aharoni et al.,
1998; Duda
et al., 2000). These cells cross the blood brain barrier (BBB), accumulate in
the CNS
(Aharoni et al., 2000, 2002), and express in situ interIeukin-10 (IL-10),
transforming
growth factor-r3 (TGF-P), as well as Brain Derived Neurotrophic Factor (BDNF)
(Aharoni et al., 2003). Furthermore, the GA-specific cells induce bystander
effect on
neighboring CNS cells to express these beneficial factors and reduce
interferon (IFN)- y
expression. A key issue in the capability of GA to counteract the pathological
process is
its effect on the neuronal system, which is the actual target of the
pathological process.
None of the above-mentioned references discloses specifically that GA induces
neurogenesis in the CNS and no data nor protocol is disclosed for testing GA
effect in the
induction of neurogenesis in the CNS.
SUMMARY OF THE INVENTION
The present invention relates to the use of a neuroprotective agent selected
from
the group consisting of Copolymer 1, a Copolymer 1-related polypeptide, a
Copolymer 1-
related peptide, and activated T cells which have been activated by Copolymer
1, a
Copolymer 1-related polypeptide, or a Copolymer 1-related peptide, for
inducing and
enhancing neurogenesis and/or oligodendrogenesis from endogenous as well as
from
exogenous stem cells administered to a patient.
In a preferred embodiment, the agent is Copolymer 1 for use in combination
with
stem cell therapy.
9
_ ¨ _

CA 02588908 2010-12-07
=
BRIEF DESCRIPTION OF THE FIGURES
Figs. 1A-1F show that differentiation of NPCs into neurons can be either
induced or
blocked by microglia, depending on how they are activated. Green fluorescent
protein
(GFP)-expressing NPCs (green) were co-cultured with differently activated
microglia
from mice for 5 days. Quantification of P-III-tubulin+ cells (expressed as a
percentage of
GFP+ cells) obtained from confocal images, without (-Ins) or with insulin
(+Ins), is
summarized in Figs. lA and 1B, respectively. Fig. 1C shows results of the
effect of
rTNF-a on the number of p-III-tubulin+ cells, expressed as a percentage of
GFP+ cells, in
co-cultures of NPCs and MG(IFN.7) in the presence of insulin. Error bars
represent means
SD. Data are from one of at least three independent experiments in replicate
cultures.
Asterisks above bars express differences relative to untreated (Control) NPCs
(* P ( 0.05;
P < 0.001; ANOVA). Fig. 1D shows representative confocal images of GFP-
expressing NPCs (green), in the absence of insulin without microglia
(Control); with
untreated microglia (MG(_)); with LPS-activated microglia (MG(Lps)); with IL-4-
activated
microglia (MG(1L_4)); and in the presence of insulin with IFN-y-activated
microglia
(MG(IFN_y)+Ins) or IFNI-activated microglia (MG(IFN1)+Ins) and aTNF. Fig. lE
shows
GFP-expressing NPCs co-expressing P-III-tubulin and Nestin. Fig. IF shows

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
that newly formed neurons from NPCs are positively stained for glutamic acid
decarboxylase (GAD) 67 (P-III-tubulin+/GFP+/GAD+). Note, confocal channels are

presented separately.
Figs. 2A-2D show that microglia activated with IFN-y or IL-4 induce
differentiation of NPCs into doublecortin (DCX)-expressing neurons with
different
morphology. GFP-expressing NPCs (green) were co-cultured with differently
activated microglia as described in Fig. 1, and stained for the neuronal
marker
DCX. Fig. 2A depicts two representative confocal images of GFP-expressing NPCs

(green) co-cultured for 5 days with MG(1L4) in the absence of insulin (left
panel) or
with MG(I) in the presence of insulin (MG(IFNI),Ins, right panel). Fig. 2B
shows
representative confocal images of GFP-expressing NPCs co-expressing DCX. Fig.
2C shows representative confocal images of P-III-tubulin+ cells co-expressing
DCX. Note, confocal channels are presented separately. Fig. 2D shows
quantification of DCX+ cells (expressed as a percentage of GFP+ cells)
obtained
from confocal images, without or with insulin. Error bars represent means SD.
Data are from one of at least three independent experiments in replicate
cultures.
Asterisks above bars express differences relative to untreated (control) NPCs
(* P <
0.05; ** P < 0.01; *** P < 0.001; ANOVA).
Figs. 3A-3E show that differentiation of NPCs into oligodendrocytes can be
either induced or blocked by microglia, depending on how they are activated.
GFP-
expressing NPCs (green) were cultured alone (Control) or co-cultured with
differently activated microglia as described in Fig. 1. Histograms showing
quantification of NG2+ or RIP+ cells (expressed as a percentage of GFP+ cells)

obtained from confocal images, co-cultures after 5 days (3A) in insulin-free
medium (-Ins) or (3B) in insulin-containing medium (+Ins). The data shown are
from one of three independent experiments in replicate cultures, with bars
representing means SD. Asterisks above bars express differences relative to
untreated (Control) NPCs (** P ( 0.01; *** P < 0.001; ANOVA). Fig. 3C shows 4
representative confocal images of GFP-expressing NPCs (green) and NG2+ (red)
cells: without microglia (Control); co-cultured with untreated microglia
(MG(.)); co-
11

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
cultured with IFN-y-activated microglia in the presence of insulin
(MG(IIN_?)+Ins);
co-cultured with IL-4-activated microglia (MG(1L_4)), for 5 days. Fig. 3D
shows
confocal images showing co-localization of GFP, NG2 and Nestin cells. Note,
confocal channels are presented separately. Fig. 3E shows that NG2+ cells are
seen
adjacent to MAC1+ cells.
Figs. 4A-4G show differentiation and maturation of NPCs in the presence of
MG(Jl) or MG(IL.4) after 10 days in culture. Cultures of untreated NPCs
(Control)
or of NPCs co-cultured with MG(JFN-y) or MG(1L4) were analyzed after 10 days.
Fig.
4A depict the numbers of NG2, RIP+, Ga1C+, GFAP+ or 13-III-tubu1in+ cells
expressed as percentages of GFP+ cells. Values are means SD (* P < 0.05; **
P <
0.01; *** P < 0.001; ANOVA). Figs. 4B-4F are representative confocal images of
NPCs in the presence of MG(IL-4) after 10 days in culture. Fig. 4B shows that
increased branching of processes stained with NG2 was seen after 10 days
(compare
Fig. 4B with Fig. 3C, MG(1L.4)). Contact is seen to be formed between an NG2+
process and an adjacent cell (high magnification of boxed area). Fig. 4C shows
that
staining of the same cultures for mature oligodendrocytes (GalC) and neurons
(r3-
III-tubu1in+) shows contacts between neurons and highly branched
oligodendrocytes
(high magnification of boxed area). Fig. 4D shows that no overlapping is seen
between labeling for neurons (DCX+) and for oligodendrocytes (RIP). Figs. 4E
and
4F show that no overlapping is seen between GFAP and NG2 labeling or between
GFAP and DCX labeling, respectively. Fig. 4G shows neurites length of MG(J)
and MG(1L4) cells. Values are means SD (*** P ( 0.001; ANOVA).
Figs. 5A-5D show the role of IGF-I and TNF-a in induction of
oligodendrogenesis by IL-4- and IFN-y-activated microglia. (5A) GFP-expressing
NPCs (green) were cultured alone (control), in the presence of aIGF-I, in co-
cultures with MG(4) in the absence or presence of aIGF-I (5 g/m1), or in the
presence of aTNF-a (1 ng/ml). (5B) In an independent experiment, NPCs were
cultured in the presence of rIGF-I (500 ng/m1). In Figs. 5A and 5B no insulin
was
added to the media. (5C) NPCs were cultured alone (control), with 5:11\1F-a (1
12

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
ng/ml), or with MG(IF) in the absence or presence of a'TNF-a. (5D) In an
independent experiment, NPCs were cultured with MG0FN-y) in the presence of
insulin and rTNF-a (10 ng/ml). Error bars represent means SD. Asterisks
above
bars express differences relative to untreated (control) NPCs (* P < 0.05; **
P <
0.01; *** P < 0.001; ANOVA).
Figs. 6A-6C show that IFN-y, unlike IL-4, transiently induced TNF-oc and
reduced IGF-I expression in microglia. (6A) Microglia treated with IL-4 (10
ng/ml),
IFN-y (20 ng/ml), or LPS (100 ng/ml) for 24 h were analyzed for TNF-a and IGF-
I
mRNA by semi-quantitative RT¨PCR. Representative results of one of three
independent experiments are shown. (6B) Time courses of TNF-a and IGF-I
mRNA expression by MG(1L4) and MG(IFN1). PCR at each time point was performed
with the same reverse-transcription mixtures for all cDNA species. Values
represent
relative amounts of amplified mRNA normalized against I3-actin in the same
sample, and are represented as fold of induction relative to control (means
SD).
The linear working range of amplifications was ascertained before the
experiments
were carried out. Each sample was tested in three replicates, and similar
results
were obtained in three different microglial cultures. (6C) Statistical
analysis of IGF-
I expression demonstrates fluorescence intensity per cell, calculated as a
percentage
of increased intensity relative to MG(_) (control) (means SD; obtained in
two
independent experiments, each repeated four times). Note, relative to the
untreated
control, MG(1L..4) showed a significant increase in IGF-I. Asterisks above bar
express
differences relative to MG(_) (* P < 0.05; ** P < 0.001; two-tailed Student's
t-test).
Figs. 7A-7H show that a myelin-specific autoimmune response operates
synergistically with transplanted aNPC transplantation in promoting functional
recovery from spinal cord injury (SCI). Recovery of motor function after SCI
(200
kdynes for 1 s) in male C57B1/6J mice (n ¨ 6-9 in each group). (7A) Mice were
immunized with MOG peptide or PBS emulsified in CFA containing 1%
Mycobacterium tuberculosis (MOG-CFA and PBS-CFA, respectively). One week
after SCI, aNPCs were transplanted into their lateral ventricles (MOG-CFA/aNPC
13

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
or PBS-CFA/aNPC). The lateral ventricles of mice in similarly injured and
immunized control groups were treated with PBS (MOG-CFA/PBS or PBS-
CFA/PBS). Values of the Basso motor score (BMS) rating scale are presented.
(7B)
BMS scores of individual mice described in (7A) on day 28 of the experiment.
(7C)
Recovery of motor function after SCI (200 kdynes for 1 s) in male C57B1/6J
mice
(n ¨ 6-9 in each group) immunized with MOG peptide 45D emulsified in CFA
containing 2.5%Mycobacterium tuberculosis. One week after SCI, aNPCs were
transplanted into the lateral ventricles. Similarly injured and immunized
control
groups, instead of being transplanted with aNPCs, were injected with PBS. BMS
values are presented. (7D) Recorded BMS scores of individual mice described in
(7C) on day 28 of the experiment. (7E) Injury and aNPC transplantation were as
in
(7A), but immunization was carried out 7 days prior to SCI and the mice were
immunized with MOG-IFA or injected with PBS (control). (7F) Injury and aNPC
transplantation were as in (7A), but immunization was carried out 7 days prior
to
SCI and the mice were immunized with OVA/CFA. (7G, 7H) Injury and aNPC
transplantation were as in (7A), but immunization was carried out 7 days prior
to
SCI and the mice were immunized with MOG peptide and CFA containing 2.5%
Mycobacterium tuberculosis. Results in all groups are means SEM. Asterisks
show differences at the indicated time points, analyzed by two-tailed
Student's t-
test. (*,p< 0.05; **,p< 0.01; ***,p< 0.001).
Figs. 8A-8F show that GFP-labeled aNPCs are found in the parenchyma of
the spinal cord after dual treatment with MOG immunization and aNPC
transplantation. Immunohistochemical staining of longitudinal paraffin
sections of
spinal cords excised 7 or 60 days after transplantation of aNPCs to the
lateral
ventricles. Sections were stained with anti-GFP antibody and counterstained
with
Hoechst to detect nuclei. They were then scanned by fluorescence microscopy
for
the presence of GFP+ cells. Representative micrographs of GFP-immunolabeled
cells in areas adjacent to the lesion site 7 days after transplantation (8A-
8F) and 60
days after transplantation of aNPCs (8A-8F) are shown.
14

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Figs. 9A-9F show histological analysis of spinal cords from injured
C57B1/6J mice after dual treatment with MOG/CFA immunization and aNPC
transplantation. Spinal cords were excised 1 week after cell transplantation.
SCI
C57B1/6J mice (n = 3-4 in each group) were subjected to SCI (200 kdynes for 1
s)
and immunized on the day of SCI with MOG peptide emulsified in CFA containing
1% Mycobacterium tuberculosis. One week after SCI, the lateral ventricles of
MOG-CFA-immunized mice transplanted aNPCs or injected PBS. (9A, 9B), GFAP
staining of longitudinal sections of injured spinal cords shows significantly
smaller
areas of scar tissue after treatment with MOG-CFA/aNPC than in any of the
other
groups. (9A) Representative micrographs of spinal cords from mice treated with
MOG-CFA/aNPC, MOG-CFA/PBS, PBS-CFA/aNPC, or PBS-CFA/PBS are
shown. (9B) Quantification of the area delineated by GFAP staining (* p (0.05,
**
p <0.01, *** p <0.001, two-tailed Student's t-test; n = 4 analyzed slices from
each
mouse). (9C, 9D) Longitudinal paraffin sections of spinal cords excised and
stained
for IB4 7 days after cell transplantation and 14 days after contusive SCI show
significantly less staining in MOG/CFA/aNPC-treated mice than in any of the
other
groups. (9D) Quantification of area occupied by IB4 staining (* p <0.05, ** p
<0.01
***p <0.001, two-tailed Student's t-test). (9E, 9F) Staining with anti-CD3
antibody
to identify infiltrating T cells at the site of injury. (9E) Representative
micrographs
of spinal cords from mice treated with MOG-CFA/aNPC, MOG-CFA/PBS, PBS-
CFA/aNPC, or PBS-CFA/PBS. Manual counting of cells in four slices from each
mouse, obtained from four areas surrounding the site of injury, disclosed
significantly more CD3+ cells in the group treated with MOG-CFA/aNPC than in
any of the other groups (* p <0.05, ** p <0.01 ***p <0.001, two-tailed
Student's t-
test).
Figs. 10A-100 show histological analysis of BDNF and noggin expression
in spinal cords from injured C57B1/6J mice after dual treatment with MOG/CFA
immunization and aNPC transplantation. C57B1/6J mice were subjected to SCI (n
=
3-4 in each group) and immunized with pMOG 35-55 emulsified in CFA (1%
Mycobacterium tuberculosis) on the day of SCI. One week after SCI, the lateral

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
ventricles of MOG-CFA-immunized mice transplanted with aNPCs or injected
PBS. Longitudinal sections of spinal cords excised 7 days after cell
transplantation
and 14 days after SCI (n ¨ 3-4 in each group) were stained for BDNF. (10A)
Quantification of area stained for BDNF (* p <0.05, ** p <0.01, ***p <0.001,
two-
tailed Student's t-test). Staining for BDNF is significantly more intense in
mice
treated with MOG-CFA/aNPC than in any of the other groups. (10B) Double
staining for BDNF and 1B4 shows that 1B4+ microglia/macrophages are a major
source of BDNF. (10C) Significantly more intense staining for noggin was found
in
mice treated with MOG/CFA/aNPCs than in any of the other groups. (10D)
Quantification of area stained with noggin (* p <0.05, ** p <0.01 ***, p
<0.001,
two-tailed Student's t-test). Double staining for noggin and IB4 shows that
IB4+
microglia are a major source of noggin.
Figs. 11A-11E show increase in BrdU/DCX double staining in the vicinity
of the site of injury after dual treatment with immunization and aNPC
transplantation. SCI and aNPCs transplantation as in Fig. 7. One week after
aNPCs
transplantation mice were injected twice daily for 3 days with BrdU.
Longitudinal
sections of spinal cords excised 14 days after cell transplantation and 28
days after
contusive SCI (n = 3-4 in each group) were stained for BrdU and DCX.
Significantly more BrdU+/DCX+ cells were found in mice treated with MOG-
CFA/aNPC than in any of the other groups.
Figs. 12A-12F show that T cells induce neuronal differentiation from aNPCs
in vitro. (Fig. 12A) Quantification of P-III-tubulin+ cells (expressed as a
percentage
of DAPI cells) after 5 days in culture alone (control), or in co-culture with
pre-
activated CD4+ T cells, or with resting CD4+ T cells (** p < 0.01; ***p <
0.001;
ANOVA). (Fig. 12B) Representative images showing 13-III-tubulin expression in
aNPCs after 5 days in culture alone (control), or in co-culture with pre-
activated
CD4+ T cells. (Fig. 12C) Quantification of P-III-tubulin+ cells (expressed as
a
percentage of DAPI cells) after 5 days in culture of aNPCs in the presence of
medium conditioned by activated T cells. (Fig. 12D) Representative images
showing branched, elongating P-III-tubulin lebeled fibers. (Fig. 12E)
Quantification
16

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
of 13-III-tubu1in+ cells in aNPCs after 5 days in culture with different
concentrations
of IFN-y or IL-4 (***p < 0.001; ANOVA). (Fig. 12F) Quantitative RT¨PCR
showing a fivefold reduction in Hes-5 expression in aNPCs cultured with medium

conditioned for 24 h by activated CD4+ T-cells.
Figs. 13A-13C show that aNPCs inhibit T-cell proliferation and modulate
cytokine production. (Fig. 13A) Proliferation was assayed 96 h after
activation by
incorporation of [311]-thymidine into CD4+ T cells co-cultured with aNPCs.
Recorded values are from one of three representative experiments and are
expressed
as means + SD of four replicates. (Fig. 13B) Proliferation of CD4+ T cells
cultured
alone, or in the presence of aNPCs (co-culture), or with aNPCs in the upper
chamber of a transwell. (Fig. 13C) Cytokine concentrations (pg/ml) in the
growth
medium 72 h after activation of CD4+ T cells alone or in co-culture with
aNPCs.
Figs. 14A-14C show that glatiramer acetate (GA) vaccination counteracts
cognitive loss in the APP/PS1 Tg mouse model of Alzheimer's disease (AD).
Hippocampal-dependent cognitive activity was tested in the MWM. (Figs. 14A-
14C) GA-vaccinated Tg mice (diamond; n=6) showed significantly better
learning/memory ability than untreated Tg mice (square; n=7) during the
acquisition and reversal phases but not the extinction phase of the test.
Untreated Tg
mice showed consistent and long-lasting impairments in spatial memory tasks.
In
contrast, performance of the MWM test by the GA-vaccinated Tg mice was rather
similar, on average, to that of their age-matched naïve non-Tg littermates
(triangle;
n = 6) (3-way ANOVA, repeated measures: groups, df (2,16), F= 22.3, P <
0.0002;
trials, df (3,48), F¨ 67.9, P < 0.0001; days, df (3,48), F = 3.1, P < 0.035,
for the
acquisition phase; and groups, df (2,16), F = 14.9, P < 0.0003; trials, df
(3,48), F =
21.7, P < 0.0001; days, df (1,16), F = 16.9, P < 0.0008, for the reversal
phase).
Figs 15A-15J show that T cell-based vaccination with GA leads to a
reduction in [3-amy1oid (A13) and counteracts hippocampal neuronal loss in the

brains of Tg mice: key role of microglia. (Fig. 15A) Representative confocal
microscopic images of brain hippocampal slices from non-Tg, untreated-Tg, and
GA-vaccinated Tg littermates stained for NeuN (mature neurons) and human A.
17

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
The non-Tg mouse shows no staining for human AP. The untreated Tg mouse
shows an abundance of extracellular AP plaques, whereas in the GA-vaccinated
Tg
mouse AP-immunoreactivity is low. Weak NeuN+ staining is seen in the
hippocampal CA1 and DG regions of the untreated Tg mouse relative to its non-
Tg
littermate, whereas NeuN+ staining in the GA-vaccinated Tg mouse is almost
normal. (Fig. 15B) Staining for activated microglia using anti-CD1lb
antibodies.
Images at low and high magnification show a high incidence of cells double-
immunostained for AP and CD1 lb in the CA1 and DG regions of the hippocampus
of an untreated Tg mouse, but only a minor presence of CD1 lb+ microglia in
the
GA-vaccinated Tg mouse. Arrows indicate areas of high magnification, shown
below. (Fig. 15C) CD1113+ microglia, associated with an AP-plaque, expressing
high levels of TNF-a in an untreated Tg mouse. (Fig. 15D) Staining for MHC-II
(a
marker of antigen presentation) in a cryosection taken from a GA-vaccinated Tg

mouse in an area that stained positively for AP shows a high incidence of MEC-
IT+
microglia and almost no TNF-a+ microglia. (Fig. 15E) MHC-Ir microglia in the
GA-vaccinated mouse co-express IGF-I. (Fig. 15F) CD3+ T cells are seen in
close
proximity to MHC-II+ microglia associated with AP-immunoreactivity. Boxed area

shows high magnification of an immunological synapse between a T cell (CD3+)
and a microglial cell expressing MHC-H. (Fig. 15G) Histogram showing the total
number of AP¨plaques (in a 30- m hippocampal slice). (Fig. 15H) Histogram
showing the total stained AP¨immunooreactive cells. Note, the significant
differences between GA-vaccinated and untreated Tg mice, and verifies the
decreased presence of AP-plaques in the vaccinated Tg mice. (Fig. 15I)
Histogram
showing a remarkable reduction in cells stained for CD1 lb, indicative of
activated
microglia and inflammation, in the GA-vaccinated Tg mice relative to untreated
Tg
mice. Note the increase in CD11b+ microglia with age in the non-Tg
littermates.
(Fig. 15J) Histogram showing increased survival rate of Neul\r- neurons in the
DGs
of GA-vaccinated Tg mice relative to untreated Tg mice. Error bars indicate
means
SEM. Asterisks above bars express the significance of differences in the
18

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
immunostaining (* P < 0.05; **p < 0.01; *** P ( 0.001; two-tailed Student's t-
test).
Note, all the mice in this study were included in the analysis (6-8 sections
per
mouse).
Figs. 16A-16E show enhanced cell renewal induced by T cell-based
vaccination with glatiramer acetate (GA) in the hippocampus of adult Tg mice.
Three weeks after the first GA vaccination, mice in each experimental group
were
injected i.p. with BrdU- twice daily for 2.5 days. Three weeks after the last
injection
their brains were excised and the hippocampi analyzed for BrdU, DCX, and NeuN.

(Fig. 16A-16C) Histograms showing quantification of the proliferating cells
(BrdU+) (Fig. 16A), newly formed mature neurons (BrdU+/NeuN+) (Fig. 16B), and
all pre-mature (DCX+-stained) neurons (Fig. 16C). Numbers of BrdU+,
BrdU+/NeuN+, and DCX+ cells per DG, calculated from six equally spaced coronal

sections (30 (..tm) from both sides of the brains of all the mice tested in
this study.
Error bars represent means + SEM. Asterisks above bars denote the significance
of
differences relative to non-Tg littermates (** P < 0.01; P <
0.001; two-tailed
Student's t-test). Horizontal lines with P values above them show differences
between the indicated groups (ANOVA). (Fig. 16D) Representative confocal
microscopic images of the DG showing immunostaining for BrdU/DCX/NeuN in a
GA-vaccinated Tg mouse and in a non-Tg littermate relative to that in an
untreated
Tg mouse. (Fig. 16E) Branched DCX+ cells are found near MHC-II+ microglia
located in the subgranular zone of the hippocampal DG of a GA-vaccinated Tg
mouse.
Figs. 17A-17D show that IL-4 can counteract the adverse effect of
aggregated A(3 on microglial toxicity and promotion of neurogenesis. (Fig.
17A) In-
vitro treatment paradigm. (Fig. 17B) Representative confocal images of NPCs
expressing GFP and 13-III-tubulin, co-cultured for 10 days without microglia
(control), or with untreated microglia, or with microglia that were pre-
activated with
AP(1-40) (5 1-11\4) (MG(Ap1-4o)) for 48 h and subsequently activated with IFN-
7 (10
ng/ml) (MG(Ap 1-40 / IFI\ly,lOng/m1)), or with IL-4 (10 ng/ml) (MG(A01-40 t IL-
4), or with both
IFNI (10 ng/ml) and IL-4 (10 ng/ml) (MG(A0 1-40 / IFNy-FIL-4)) = Note,
aggregated Af3
19

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
induces microglia to adopt an ameboid-like morphology, but after IL-4 was
added
these microglia exhibited a ramified-like structure. (Fig. 17C) Separate
confocal
images of NPCs co-expressing GFP and 13-III-tubu1in adjacent to CD11b+
microglia. (Fig. 17D) Quantification of cells double-labeled with GFP and 13-
III-
tubulin (expressed as a percentage of GFP+ cells) obtained from confocal
images.
Results are of three independent experiments in replicate cultures; bars
represent
means SEM. Asterisks above bars denote the significance of differences
relative
to untreated (control) NPCs P < 0.05; *** P < 0.001; two-tailed Student's t-
test).
Horizontal lines with P values above them show differences between the
indicated
groups (ANOVA).
Fig. 18 shows the effect of the administration of stem cells in combination
with glatiramer acetate to a mice model of amyotrophic lateral sclerosis
(ALS).
Figs. 19A-19B show clinical manifestations of EAE induced by MOG
peptide 35-55. (Fig. 19A) In C57BL/6 and YFP2.2 mice. (Fig. 19B) The effect of
GA treatment in C57BL/6 mice treated by 5-8 daily injections of GA in
different
stages of the disease i.e. starting immediately after disease induction -
prevention
treatment, starting after the appearance of disease manifestations at day 20 -

suppression treatment, or during the chronic phase 6 weeks after disease
appearance
- delayed suppression. The injection period of each treatment is illustrated
along the
x axis (n=6).
Figs. 20A-20E show histological manifestations of EAE induced by MOG
peptide 35-55. (Figs. 20A-20D) The effect of GA treatment in sagital brain
sections
of YFP2.2 mice expressing YFP (green) on their neuronal population: (Fig. 20A)

Deterioration and transaction of YFP expressing fibers in the cerebellum and
correlation with perivascular infiltration. Inserts indicate area with
perivascular
infiltrations, demonstrated by staining with antibodies for the T-cell marker
¨ CD3.
(Fig. 20B) Elimination of fibers in lesions in the striatum. (Fig. 20C)
Typical
morphology of pyramidal cells in layer 5 of the cerebral cortex. Arrows and
insert
indicate abnormal neuronal cell bodies with marginalized nuclei in EAE mice.
Considerably less damages were found in brains of EAE+GA mice than in brains
of

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
untreated EAE mice i.e. less deteriorating fibers, reduced number of lesions
with
smaller magnitude, and less swollen cell nuclei. Note the thin layer of YFP
positive
fiber, frequently found over the lesions in GA treated mice. (Fig. 20D)
Staining
with Fluoro-Jade B (green), which binds to degenerating neurons, in the cortex
of
C57BL/6 mice, 25 days after disease induction. Scale bar indicates: 500 pm in
(Fig.
20A) 50 1.1m in (Figs. 20B, 20C) and 20 pm in (Fig. 20E). L-2, L-5 and L-6,
layer
two, five and six of the cerebral cortex.
Figs. 21A-21B show microglial activation in EAE YFP2.2 mice. (Fig. 21A)
Correlation of the expression of the microglia and macrophage marker MAC-1
(red)
with deterioration and injury of YFP expressing fiber (green) in the white
matter of
the cerebellum. In box I, highly activated microglia cells are observed,
accompanied
by reduction in fiber density, whereas, in nearby area in box II low MAC-1
expression and normal fiber appearance are present. (Fig. 21B) The effect of
GA
on MAC-1 expression and on microglial cell morphology in various brain regions
of EAE mice: striatum, thalamus (dorsal lateral geniculate nucleus) and
hippocampus (granular and molecular layers). Increased MAC-1 staining and cell

morphology typical for activated microglia were displayed in brains of EAE
mice
(inserts). In contrast, MAC-1 expression in brains of EAE+GA mice was
extensively reduced exhibiting cell morphology similar to that of unactivated
microglia in naive mice. EAE was induced in YFP2.2 mice, 35 days before
perfusion. GA treatment was applied by 8 daily injections, starting
immediately
after EAE induction. Sagital sections. Scale bar indicates: (Fig. 21A) 500 pm,
(Fig.
21B) 100 pm in the striatum and thalamus, 5011m in the hippocampus.
Figs. 22A-22E show proliferation of newly generated neurons visualized by
immunostaining for the proliferation marker BrdU (red) and the immature
neuronal
marker DCX (green) in the neuroproliferative zones of C57BL/6 mice. Increased
expression of BrdU and DCX in EAE mice and to a greater extent in EAE+GA
mice, (Fig. 22A) in the SVZ, confocal images and (Fig. 22C) in the hippocampal

SGZ, 25 days after EAE induction, 1 day after last GA injection. Note the DCX+
cells in the hippocumpus that migrated into the GCL and manifest dense and
21

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
branched dendritic tree. (Fig. 22B) DCX expression in the SVZ at different
times
points: 1 day (I), 10 (II) and 30 (III) days after the last GA injection.
Neuroproliferation declined with time, still, DCX expression in GA treated
mice
was higher than in EAE mice, 1 and 10 days after treatment. Coronal sections.
Scale
bar indicates: 50 jam in (Fig. 22A), 200 [tm in (Fig. 22B) and (Fig. 22C), and
20 pin
in the right panels of (Fig. 22C). st, striatum; LV, lateral ventricle; SGZ,
subgranular zone; GCL, granular cell layer; IML, OML, inner and outer
molecular
layer. (Fig. 22D) Quantitative analysis of BrdU incorporation and DCX
expression
in EAE (red) and EAE+GA (blue) mice, at various time points after EAE
induction
and GA treatment. Increased neuronal proliferation is observed in both
neuroproliferative zones following disease appearance; subsequent decline
below
that of naive mice, and augmentation of neuroproliferation by the various
schedules
of GA treatment. Quantification was performed in the SVZ by counting BrdU
positive cells, (those with BrdU/DCX dual staining), and measuring the DCX
stained area, starting at the level of the medial septum and 640 pm backward,
and in
the hippocampal DG by counting BrdU+/DCX+ cells (in both blades), and DCX
cells (in the upper blade of the dentete), through its septo-temporal axis.
The
number of BrdU/DCX stained cells for each brain structure was averaged from 8
unilateral levels per mouse, 80 pm apart, 3-4 mice for treatment group.
Results are
expressed as change fold from naive controls. Control values for BrdU
incorporation: in the SVZ 211+31 and 23+6, in the hippocampus 45+13 and 17+8,
BrdU/DCX+ cells, one day and one month after the last BrdU injection
respectively;
for DCX staining: in the SVZ 19,464+3550 m2 and in the hippocampus 78+12 m2
positive cells averaged from 10 naïve mice. Statistical analysis was performed
by
ANOVA followed by Fisher's LSD when appropriate. * significant effect over
naïve control, # significant effect over EAE untreated mice, (p <0.05). (Fig.
22E)
Schedule of experiments: time length from EAE induction (day 0) till
perfusion;
GA injections as prevention (P), suppression (S) or delayed suppression (DS)
treatments, and BrdU inoculation - concurrently or immediately following GA
treatment.
22

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Figs. 23A-23H show promoted mobilization and migration of neuronal
progenitor cells in EAE mice treated with GA through migratory streams. (Fig.
23A) Schematic sagital representation of the migratory routes from the
subventricular zone (SVZ) through both the rostra' migratory stream (RMS - in
red)
and the lateral cortical stream (LCS - in yellow). (Fig. 23B) Sagital section
through
the RMS showing the route of DCX-positive cells (red) from the SVZ to the OB.
(Fig. 23C) neuroprogenitors in the LCS, generally functional in the embryonic
forebrain, and reappear after GA treatment in EAE adult mice. DCX-positive
cells
(red) migrate alongside the YFP expressing fibers (green) of the interface
between
the hippocampus and the corpus callosum, towards various cortical regions
mainly
in the occipital cortex. (Figs. 23D-23E) Increased mobilization of newly
generated
neurons visualized with BrdU (orange) and DCX (green) immunostaining, in the
RMS of EAE+GA mice, in comparison to EAE mice and naïve controls, in RMS
segment adjacent to the SVZ (Fig. 23D) and in a more medial section of the RMS
arc (Fig. 23E). Sagital sections. Scale bar indicates: 1000 um in (Fig. 23B),
25 in
(Fig. 23C), 500 in and (Fig. 23D), 50 um in (Fig. 23E). LV, lateral ventricle;
Ctx,
cortex; St, striatum; OB, olfactory bulb; AC, anterior commissure; cc, corpus
callossum; Hip, hippocampus; L-5 and L-6, layer five and six of the cerebral
cortex.
(Figs. 23F-23H), Quantitative analysis of BrdU (co-expressing DCX) or DCX in
the
RMS, one day (Figs. 23F, 23G), and one month (Fig. 23H) after termination of
BrdU and GA injections, indicating significant increase of neuroprogenitors in
the
RMS of EAE mice over control, and higher elevation in EAE+GA mice. Note that
in one EAE mouse which exhibited slight, short-term disease and spontaneous
recovery (EAE-rec, Fig. 23H), enhanced neuronal migration was observed.
Quantification was performed by counting the BrdU/DCX+ cells and measuring the
DCX stained area (in 0.22 mm), along the striatal border. The amount of
BrdU/DCX
stained cells was averaged from 8 sections per mouse, 80 um apart. Three mice
counted per treatment group, except EAE rec, which shows a single mouse.
Results
are expressed as change fold from naive controls. Control values for BrdU
incorporation: 146+31 BrdU/DCX cells, one day after the last BrdU injection,
and
23

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
for DCX staining: 2193+305 m2, averaged from 6 naïve mice. *p<0.05 versus
naïve control. EAE mice in (Figs. 23B, 23C, 23H) were treated with GA
subsequent
to disease induction, one month before perfusion - prevention, in (Figs. 23D,
23E
and 23F, 23G) EAE induced mice were injected with GA and BrdU 20 days post
disease induction, 1-5 days before perfusion - suppression.
Figs. 24A-24F show migration of neuronal progenitor cells in EAE induced
mice treated with GA. DCX expressing neuronal progenitors (orange) diverge
from
the classic neuroproliferative zones or the migratory streams and spread to
atypical
regions along YFP expressing fibers (green). (Fig. 24A) From the RMS into the
in
the striatum. (Figs. 24B, 24C) Towards the region of the nucleus accumbens,
from
the SVZ (Fig. 24B) and from the RMS (Fig. 24C). (Fig. 24D) From the RMS into
the internal part of the cortex to ¨ (Fig. 24E), layer 5, and (Fig. 24F),
layer 6. Note
the morphological features of the DCX expressing cells - fusiform somata with
a
leading and trailing processes (Fig. 24C insert and Figs. 24E, 24F),
characteristic of
migrating neurons, and their orientation - migration away from the migratory
stream, along the nerve fibers (Figs. 24A 24D-24F). Sagital sections. Scale
bar
indicates: 200 jum in Figs, 24A-24D, 100 lam in Fig. 24E and 10 gm in Fig.
24F. In
Figs. 24A-24C enlarged box area is depicted in the right panel. RMS, rostral
migratory stream; SVZ, subventricular zone; St, striatum; AC, anterior
commissure;
cc, corpus callossum; L-5 and L-6, layer five and six of the cerebral cortex.
Figs. 25A-25K show Fate tracing of neuronal progenitor cells generated in
the course of GA treatment in EAE mice. BrdU incorporated cells (red), born
during
the concurrent injections of BrdU and GA migrated to various brain regions and

expressed neuronal markers. (Figs. 25A, 25B) BrdU positive cells co-expressing
the
immature neuronal marker DCX (green), 10 days after the last injection, in the
striatum (Fig. 25A), and in the accombens nucleus (Fig. 25B). Note the
clusters of
double positive cells suggesting local divisions. (Fig. 25C) Staining of DCX
expressing cells in the accombens nucleus with the endogenous proliferation
marker
phosphohiston (blue), showing DCX positive cells that had proliferated in situ
prior
to sacrifice of the mouse. (Figs. 25D-25G) BrdU positive cells co-expressing
the
24

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
mature neuronal marker NeuN (green), one month after completion of GA/BrdU
injections, in the striatum (Fig. 25D, 25F), in the nucleus accumbens (Fig.
25E), and
in the cingulate cortex layer 5 confocal image (Fig. 25G). A1TOWS indicate
representative BrdU/NeuN co-expressing cells. (Figs. 25H, 25K) BrdU positive
cells, one month after GA/BrdU injection in YFP mice, co-expressing YFP
(green)
in the cingulate layer 5 (Figs. 25H, 251), occipital layer 6 (Fig. 254 and
motor layer
5 (Fig. 25K) of the cortex. Pyramidal cells with characteristic elongated
apical
dendrites and axons, indicative of mature functional neurons can be seen. Fig.
25G
and Fig. 25K are confocal images. Sagital sections. Scale bar indicates: 200
vim in
(Figs. 25A, 25B, 25H), 100 pm in Figs. 25C, 25D 50 vim in Figs. 25E, 25I-25K,
15
pm in Figs. 25F, 25G.
Figs. 26A-26F show migration of neuronal progenitors to lesion sites. DCX
expressing cells (orange) were found in injured regions with deterioration of
YFP
expressing fibers (green). (Fig. 26A), In EAE mice (not treated by GA), 35
days
after disease induction, in the striatum. (Figs. 26B-26F), In EAE mice, 35
days after
disease induction, treated by GA (8 daily injections, starting immediately
after
disease induction - prevention). DCX expressing cells diverging from the RMS
towards a lesion in the striatum (Fig. 26B), surrounding a lesion in the
striatum
(Fig. 26C), inside a lesion in the frontal cortex layer 5/6 (Figs. 26D, 26E),
and in a
cluster surrounding a lesion in the accumbens nucleus (Fig. 26F). Lesions in
GA-
treated mice were less extensive than in untreated mice, yet the amount of
progenitors adjoining these lesions was extensively higher. Note the YFP
expressing fibers extending into the lesions and the axonal sprouting in
lesions
occupied by DCX expressing cells (Figs. 26D-26F). Sagital sections. Scale bar
indicates: 100 pm in Figs. 26A, 26B , 50 vim in Figs. 26C, 26D, 26F, and 20
jum in
Fig. 26E.
Figs. 27A-27F show that GA induced neuronal progenitors migrate to gliotic
scar areas and express in situ BDNF. Figs. 27A-27C, DCX expressing cells
(green),
in regions populated with GFAP expressing astrocytes (red), in the striatum.
Figs.
27D-27F, DCX expressing cells (green) manifest extensive expression of BDNF

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
(orange) in the nucleus accombance (Fig. 27D, 27E) and the hippocampal dentate

gyrus (Fig. 27F). Coronal sections. Scale bar indicates: 100 pm in Fig. 27A-
27C, 50
um in Fig. 27D, 12 um in Fig. 27E and 30 um in Fig. 27F.
DETAILED DESCRIPTION OF THE INVENTION
While trying to elucidate the effect of EAE induction on neurogenesis and
differentiation towards the neural lineage and to investigate whether
peripheral
immunomodulatory treatment with GA injection in various stages of disease has
any effect on neurogenesis and neuroprotective processes, it was found by some
of
the inventors that in EAE mice neuroproliferation was elevated following
disease
appearance, but subsequently declined below that of naive mice. In contrast,
GA
treatment led to sustained reduction in the neuronal/axonal damage and
augmented
neuroprogenitor proliferation and mobilization. The newborn neuroprogenitors
manifested massive migration through exciting and dormant migration pathways,
into injury sites in brain regions, which do not normally undergo
neurogenesis, and
differentiated to mature neuronal phenotype, endorsing a direct linkage
between
immunomodulation, neurogenesis and therapeutic consequence in the CNS.
Research during the last decade has disclosed that the brain is potentially
capable of cell renewal throughout life, albeit to a limited extent (Morshead
et al.,
1994). However, the mechanisms that might restrict or favor the renewal of
adult
neural cells are not known. Recent studies from the laboratory of the present
inventors have shown that after an injury to the CNS a local immune response
that
is properly controlled in time, space, and intensity by the peripheral
adaptive
immunity is a pivotal requirement for posttraumatic neuronal survival (Moalem
et
al., 1999; Butovsky et al., 2001; Schwartz et al., 2003; Shaked et al., 2004).
We
therefore envisaged the possibility that the lack of neurogenesis and the
restricted
recovery might be attributable to a common factor, which might in turn be
related to
the local immune response.
The present invention is based on the assumption of some of the inventors
that well-regulated adaptive immunity is needed for cell renewal in the brain.
It was
26

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
thus postulated that neurogenesis and oligodendrogenesis are induced and
supported
by microglia that encounter cytokines associated with adaptive immunity, but
are
not supported by naïve microglia and are blocked by microglia that encounter
endotoxin.
In fact, it is shown herein that certain specifically activated microglia can
induce and support neural cell renewal. Thus, both neurogenesis and
oligodendrogenesis were induced and supported in NPCs co-cultured with
microglia activated by the cytokines IL-4 and IFN-y, both associated with
adaptive
immunity. In contrast, microglia exposed to LPS blocked both neurogenesis and
oligodendrogenesis, in line with previous reports that MG(LPs) block cell
renewal
(Monje et al., 2003).
Defense mechanisms in the form of activated microglia are often seen in
acute and chronic neurodegenerative conditions, and the CNS is poorly equipped
to
tolerate them (Dijkstra et al., 1992). As a result, activated microglia have
generally
been viewed as a uniformly hostile cell population that causes inflammation,
interferes with cell survival (Popovich et al., 2002), and blocks cell renewal
(Monje
et al., 2002, 2003).
Recent studies have shown, however, that the type of activation determines
microglial activity, and that just as their effects can be inimical to cell
survival in
some circumstances, they can be protective in others. Thus, for example,
microglia
that encountered adaptive immunity (CD4+ T cells) were shown to acquire a
protective phenotype (Butovsky et al., 2001). Among the cytokines that are
produced by such T cells and can endow microglia with a neuroprotective
phenotype are IFN-y and IL-4, characteristic of Thl and Th2 cells,
respectively.
Thus, microglia exposed to activated Thl cells or to IFN-y show increased
uptake of
glutamate, a key player in neurodegenerative disorders (Shaked et al.,
unpublished
observation), while their exposure to IL-4 results in down-regulation of TNF-
a, a
common player in the destructive microglial phenotype, and up-regulation of
insulin-like growth factor (IGF-1) (shown herein in the examples), which
promotes
differentiation of oligodendrocytes from multipotent adult neural progenitor
cells
27

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
(Hsieh et al., 2004). In addition, IGF-1 prevents the acute destructive effect
of
glutamate-mediated toxicity on oligodendrocytes in vitro (Ness et al., 2002)
and
inhibits apoptosis of mature oligodendrocytes during primary demyelination
(Mason et al., 2000). These and other findings strongly suggest that the
outcome of
the local immune response (in terms of its effect on the microglia) in the
damaged
CNS will be either beneficial or harmful, depending on how the microglia
interpret
the threat.
In general, tissue repair is a process that is well synchronized in time and
space, and in which immune activity is needed to clear the site of the lesion
and
create the conditions for migration, proliferation, and differentiation of
progenitor
cells for renewal. In light of the well-known fact that constitutive cell
renewal is
limited in the CNS, as well as the reported observations that treatment with
MG(LPs)
causes neuronal loss (Boje et al., 1992) and interferes with the horning and
differentiation of NPCs (Monje et al., 2003), and that adaptively activated
microglia
can support neuronal survival, it is not surprising to discover that immune
conditions favoring neuronal survival will also support cell renewal. MG(LPs)
produce excessive amounts of NO (causing oxidative stress) and TNF-a, as well
as
other cytotoxic elements, leading to a spiral of worsening neurotoxicity (Boje
et al.,
1992). NO was found to act as an important negative regulator of cell
proliferation
and neurogenesis in the adult mammalian brain (Packer et al., 2003), and TNF-a
has an inhibitory effect on oligodendrogenesis (Cammer et al., 1999).
The results of the present invention show that MG(LPs) are indeed detrimental
to NPC survival and differentiation, but that when microglia are activated by
cells
or cytokines possessing adaptive immune function, not only are they not
cytotoxic
but they even exert a positive effect on NPC proliferation, inducing and
supporting
their differentiation into neurons or oligodendrocytes. In vivo, injection of
MG(1L4)
into rat brain lateral ventricles resulted in no neuronal loss, minimal
migration of
microglia to the CNS parenchyma, and the appearance of new neurons and
oligodendrocytes (indicated by the double-staining of BrdLT+ cells with
markers of
neurons or oligodendrocytes). Staining for microglia revealed significant
invasion
28

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
of the healthy CNS by MG(Lps), with consequent massive tissue loss, unlike in
the
case of MG(IL.4) or MG(._). Interestingly, in the non-injected hippocampus,
resident
microglia were found adjacent to the subventricular zone. It is tempting to
speculate
that these might be the cells responsible for controlling neurogenesis,
restraining it
when in their resting state (as found in the present work using MGH), but
inducing
and supporting it when suitably activated.
Our findings are supported by the observation that in mice with experimental
autoimmune encephalomyelitis (EAE), NPCs migrate to sites of CNS damage
(Pluchino et al., 2003). They are also in line with the common experience that
cell
renewal is favored by injury, since they imply that in the absence of injury
the
conditions that might favor renewal do not exist.
Renewal of cells and their replenishment by new growth is the common
procedure for tissue repair in most tissues of the body. It was thought that
in the
brain those processes do not occur, and therefore that any loss of neurons,
being
irreplaceable, results in functional deficits that range from minor to
devastating.
Since an insult to the CNS, whether acute or chronic, is often followed by the

postinjury spread of neuronal damage, much research has been devoted to
finding
ways to minimize this secondary degeneration by rescuing as many neurons as
possible.
70 The
results of the present invention lead us to an intriguing conclusion. First,
under pathological conditions (when cell renewal is critical), not only do the

microglia not favor cell renewal, but they interfere with it. Secondly, this
paradoxical situation can be remedied by well-controlled adaptive immunity,
which
shapes the microglia in such a way that their activity is not cytotoxic but is
both
protective and conducive to renewal. This indicates that in those cases in
which
protective autoimmunity leads to improved recovery, both neurogenesis and
gliogenesis are likely to occur. These data can also explain the lack of cell
renewal
in autoimmune diseases; in such cases, it is likely that the quantity of
circulating
autoimmune T cells exceeds the threshold above which TNF-a production, due to
an excess of IFN-y, does not allow the microglia to acquire a protective
phenotype.
29

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
They can also explain why steroids are not helpful, as their anti-inflammatory

activity masks not only the destructive but also the beneficial adaptive
immunity.
The therapy of choice for both autoimmune diseases and neurodegenerative
conditions would therefore appear to be immunomodulation in which, after the
acute phase of disease, the surviving tissue can be maintained by relatively
small
quantities of T cells.
The findings of the present invention indicate that the limitation of
spontaneous, endogenous neurogenesis and oligodendrogenesis in the adult brain
is,
at least in part, an outcome of the local immune activity, and that harnessing
of
adaptive immunity rather than immunosuppression is the path to choose in
designing ways to promote cell renewal in the CNS.
Cell renewal in the adult mammalian CNS is limited. Recent studies suggest
that it is arrested by inflammation. That view is challenged by the findings
of the
present invention that microglia, depending on environmental stimulation, can
either induce and support or block such renewal. In vitro, neurogenesis and
oligodendrogenesis from neural progenitor cells were shown herein to be
promoted
by mouse microglia that encountered T-cell-associated cytokines (IFNI, IL-4),
but
were blocked by microglia that encountered endotoxin. Anti-IGF-1 antibodies
neutralized the IL-4 effect, while anti-TNF-a antibodies augmented the effect
of
IFN-y. Injection of IL-4-activated microglia into cerebral ventricles of adult
rats
induced significant hippocampal neurogenesis and cortical oligodendrogenesis,
whereas endotoxin-activated microglia caused neuronal loss and blocked
neurogenesis and oligodendrogenesis. These results strengthen our assumption
that
controlled adaptive immunity, unlike uncontrolled (e.g. endotoxin-induced)
inflammation, activates microglia to induce and support neuronal and
oligodendrocyte survival and renewal. Thus, to promote cell renewal in the
CNS,
well-controlled immunity is needed and should not be suppressed.
It has been reported that the controlled activity of T cells directed to auto-
antigens in the CNS is needed for postinjury survival and repair (Moalem et
al.,
1999; Yoles et a., 2001; Kipnis et al., 2002). These results led us to suspect
that a

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
fundamental role of autoimmune T cells, known to be present in healthy
individuals, is to help maintain the integrity of the CNS, and that their
remedial
effect in a neurodegenerative environment is a manifestation of the same
restorative
role under extreme conditions. Moreover, accumulating evidence attesting to
the
participation of such autoimmune T cells in postinjury neuronal survival led
us to
postulate that if they have a similar role in the healthy CNS, it might well
have to do
with neurogenesis in adult life, possibly by maintaining the conditions needed
for
such cell renewal.
According to the present invention, we examined how the nature of
microglial activation affects neurogenesis in the adult rat hippocampus under
physiological and pathological conditions associated with brain inflammation.
Transient inflammatory conditions associated with transient accumulation of
myelin-specific Th1 cells promoted neurogenesis. Injection of microglia (MG)
activated by IFN-y (MG(IFNI)) or by IL-4 (MG(IL.4)) into the lateral
ventricles of the
brains of healthy rats promoted neurogenesis. In rats that developed
monophasic
(transient) EAE, the induced neurogenesis was further promoted by MG(1L..4).
Our
results in vitro showed that MG(FN.y) supported neurogenesis from adult rat
NPCs as
long as the IFN-y concentration was low. The impediment to neurogenesis
imposed
by high-dose IFN-y could be counteracted by IL-4. Neurogenesis induced by IL-4
was weaker, however, than that induced by low-dose IFN-y or by high-dose IFN-y
administered in combination with IL-4.
We have identified herein cellular elements in the CNS that can respond to
local environmental changes and needs, and consequently can support the
formation
of new cells from adult aNPCs. We demonstrated that once the microglia become
suitably activated by circulating T cell-derived cytokines, they can induce
neuronal
and oligodendroglial differentiation from aNPCs. In view of that observation,
and
our previous demonstration in rodents that a T cell-based vaccination promotes

recovery from contusive spinal cord injury (SCI), we postulated that
translation of
those findings into a therapeutic approach might benefit the repair process by
creating a niche-like neurogenic/gliogenic environment at the injured site.
Thus, we
31

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
expected to find that supplementing the vaccination by transplantation of
homologous aNPCs would further promote functional recovery after SCI. In the
present invention we in fact demonstrated, using a mouse model, synergistic
interaction between T cell-based immune activation and transplanted aNPCs in
promoting functional motor recovery after contusive injury of the spinal cord.
Previous studies by the inventor M. Schwartz have shown that systemic
manipulations of the immune system, based on increasing the numbers of T cells

directed to weak agonists of autoantigens, beneficially affect
neurodegenerative
conditions by promoting neuronal survival (Moalem et al., 1999; Hauben et al.,
2001; Schwartz and Kipnis, 2002). The same manipulations, for example, T cell-
based vaccination, is proposed here for increasing neurogenesis, yielding
novel
ways to maintain the integrity of the aging brain and the diseased mind.
It thus seems that maintenance and repair of brain cells necessitate a dialog
between CNS-autoreactive T cells and brain-resident microglia. This dialog
cannot
take place, however, unless the microglia are able to act as APCs, presenting
the
relevant antigens to the homing T cells. We therefore postulated that in order
to halt
the progression of Alzheimer disease (AD), T cells that recognize CNS-specific

antigens other than aggregated amyloid-P (AP) must target sites of aggregated
AP
plaques in the brain. On reaching these sites they become activated by the
encounter
with their specific antigens, presented to them by microglia acting as APCs.
Such
activation enables these T cells to offset the negative effect of aggregated
Aí3 on
locally resident microglia, thus preventing the latter from becoming cytotoxic
to
neurons and blocking neurogenesis. We tested this hypothesis by vaccinating AD

mice with glatiramer acetate (GA, also known as copolymer 1 or Cop-1), a
synthetic
copolymer approved by the FDA for treatment of multiple sclerosis, and capable
of
weakly cross-reacting with a wide range of CNS-resident autoantigens (Kipnis
et
al., 2000). GA-activated T cells, after infiltrating the CNS, have the
potential to
become locally activated without risk of the overwhelming proliferation that
is
likely to cause an autoimmune disease. Studies by the present inventors and
others
32

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
have shown that GA can simulate the protective and reparative effects of
autoreactive T cells (Kipnis et al., 2000; Benner et al., 2004).
In the present invention, APP/PS1 double-transgenic AD mice (which
coexpress mutated human presenilin 1 and amyloid-I3 precursor protein)
suffering
from decline in cognition and accumulation of A13 plaques, a T cell-based
vaccination, by altering the microglial phenotype, ameliorated cognitive
performance, reduced plaque formation, rescued cortical and hippocampal
neurons,
and induced hippocampal neurogenesis.
We show here that vaccination of Tg mice with GA reduced plaque
formation, and prevented and even partially reversed cognitive decline, even
if the
vaccination was given after some loss of cognition and some plaque formation
had
already occurred. It should be noted that vaccination with GA was effective
not
only in preventing disease progression but also¨when administered after onset
of
the clinical symptoms of learning/memory loss and pathological appearance of
plaques¨in promoting tissue repair. The above findings are in line with our
observation that in mice deficient in CNS-autoreactive T cells the expression
of
brain-derived neurotrophic factor (BDNF), known to be associated with both
cognitive activity and cell renewal, is impaired. They are also in accord with
our
observation that T cells are needed for the maintenance of ccognitive
functioning in
the healthy as well as in the diseased brain (Kipnis et al., 2004). Since
aggregated
A[3 evidently interferes with the ability of microglia to engage in dialog
with T
cells, its presence in the brain can be expected to cause loss of cognitive
ability and
impairment of neurogenesis. Horning of CNS-autoreactive T cells to the site of

disease or damage in such cases is critical, but will be effective only if
those T cells
can counterbalance the destructive activity of the aggregated A[3. Myelin-
presenting
microglia, with which myelin-specific T cells can readily hold a dialog, are
likely to
be present in abundance. Myelin-related antigens, or antigens (such as GA)
that are
weakly cross-reactive with myelin, are therefore likely to be the antigens of
choice
for the therapeutic vaccination. Myelin-specific T cells will then home to the
CNS
and, upon encountering their relevant APCs there, will become locally
activated to
33

CA 02588908 2010-12-07
supply the cytokines and growth factors needed for appropriate modulation of
harmful microglia like those activated by aggregated A. The resulting synapse
between
T cells and microglia will create a supportive niche for cell renewal by
promoting
neurogenesis from the pool of adult stem cells, thereby overcoming the age-
related
impairment induced in the inflammatory brain.
The known features of glatiramer acetate (GA, Copolymer 1) are also of essence

in relation to stem cell transplantation for the treatment of neurological and
other
disorders. It is therefore proposed to use GA to improve the therapeutic
outcome of stem
cell transplantation for various disorders including neurological diseases,
especially MS.
The rationale for GA usage is for this purpose is based on previous results of
the
inventors R. Aharoni and R. Arnon concerning its mechanism of action as well
on their
findings of its efficacy in reducing graft and bone marrow rejection and self-
neurogenerating effects. It is thus envisioned that GA will be effective not
only in
augmenting endogenous neurogenesis of self-neuroprogenitor cells but also
exogenous
neurogenesis of transplanted multipotent (stem) progenitor cells. These
manifestations of
GA function taken together with its very high safety profile, support its
application in
combination therapy for the improvement of progenitor stem cell
transplantation for
many clinical applications, in addition to those specifically related to
neurological
disorders.
The present invention thus relates, in one aspect, to the use of a
neuroprotective
agent selected from Copolymer 1, a Copolymer 1-related polypeptide, a
Copolymer 1-
related peptide, and activated T cells which have been activated by Copolymer
1, a
Copolymer 1-related polypeptide, or a Copolymer 1-related peptide, for
inducing and
enhancing neurogenesis and/or oligodendrogenesis from endogenous as well as
from
exogenously administered stem cells.
34

CA 02588908 2010-12-07
The use of the invention further includes proliferation, differentiation and
survival
of newly formed neurons or oligodendrocytes, and includes neuronal progenitor
proliferation, neuronal migration, and/or neuronal differentiation of newly
formed
neurons into mature neurons.
In one embodiment, the present invention relates to inducing and enhancing
neurogenesis from endogenous or exogenously applied stem cells.
In another embodiment, the use is for inducing and augmenting self-
neurogenesis in
damaged or injured brain regions, both in brain regions that normally undergo
neurogenesis and in brain regions that normally do not undergo neurogenesis
such as
striatum, nucleus accumbens and/or cortex.
In another embodiment, the agent for use in the invention are T cells which
have
been activated by Copolymer 1, a Copolymer 1-related polypeptide, or a
Copolymer 1-
related peptide. The T cells can be endogenous and activated in vivo by
administration of
the antigen or peptide, thereby producing a population of T cells that
accumulate at a site
of injury or disease of the CNS or PNS, or the T cell are prepared from T
lymphocytes
isolated from the blood and then sensitized to the antigen. The T cells are
preferably
autologous, most preferably of the CD4 and/or CD8 phenotypes, but they may
also be
allogeneic T cells from related donors, e.g., siblings, parents, children, or
HLA-matched
or partially matched, semi-allogeneic or fully allogeneic donors. Methods for
the
preparation of said T cells are described in the above-mentioned WO 99/60021.
The invention is useful for inducing and enhancing neurogenesis and/or
oligodendrogenesis both from endogenous and exogenously administered stem
cells and
and may assist in solving the problems found today with the poor results of
stem cell
transplantation, particularly in the cases of injuries, diseases, disorders
and conditions of
the nervous system, both the CNS and PNS.
. .4

CA 02588908 2010-12-07
In one embodiment, the method of the invention is applied to induce and
enhance
neurogenesis and/or oligodendrogenesis from endogenous pools of neural
stem/progenitor cells. Thus, Copolymer 1, a Copolymer 1-related polypeptide, a
Copolymer 1-related peptide, and T cells activated therewith will by
themselves boost
endogenous neurogenesis and oligodendrogenesis in damaged tissues, supporting
also the
survival of the new neurons and oligodendrocytes.
In another embodiment, the use of the invention is applied to induce and
enhance
neurogenesis and/or oligodendrogenesis from both endogenous and exogenous stem
cells
administered to the patient. The administration of said neuroprotective agent
will assist to
enhance the successful engraftment of the implanted stem cells, cell renewal
and
differentiation of the stem cells into neurons and/or oligodendrocytes, while
at the same
time inducing the endogenous neurogenesis and oligodendrogenesis in the
damaged
tissues and supporting the survival of the new neurons and oligodendrocytes.
In one
embodiment, the use of the invention is applied to individuals suffering from
an injury,
disease, disorder or condition of the central nervous system (CNS) or
peripheral nervous
system (PNS).
The CNS or PNS njury to be treated according to the invention, preferably by
stem
cell therapy, include spinal cord injury, closed head injury, blunt trauma,
penetrating
trauma, hemorrhagic stroke, ischemic stroke, cerebral ischemia, optic nerve
injury,
myocardial infarction or injury caused by tumor excision. The transplanted
stem cells
will migrate to the region of the injury where cells had died (for example,
due to
ischaemia) and will differentiate into neurons and/or oligodendrocytes.
The CNS or PNS diseases, disorders or conditions to be treated according to
the
invention, preferably by stem cell therapy, include Parkinson's disease and
Parkinsonian
disorders, Huntington's disease, Alzheimer's disease, multiple sclerosis, or
amyotrophic
lateral sclerosis (ALS).
36

CA 02588908 2010-12-07
Other diseases, disorders or conditions include facial nerve (Bell's) palsy,
glaucoma, Alper's disease, Batten disease, Cockayne syndrome, Guillain-Barre
syndrome, Lewy body disease, Creutzfeldt-Jakob disease, or a peripheral
neuropathy
such as a mononeuropathy or polyneuropathy selected from the group consisting
of
adrenomyeloneuropathy, alcoholic neuropathy, amyloid neuropathy or
polyneuropathy,
axonal neuropathy, chronic sensory ataxic neuropathy associated with Sjogren's

syndrome, diabetic neuropathy, an entrapment neuropathy nerve compression
syndrome,
carpal tunnel syndrome, a nerve root compression that may follow cervical or
lumbar
intervertebral disc herniation, giant axonal neuropathy, hepatic neuropathy,
ischemic
neuropathy, nutritional polyneuropathy due to vitamin deficiency,
malabsorption
syndromes or alcoholism, porphyric polyneuropathy, a toxic neuropathy caused
by
organophosphates, uremic polyneuropathy, a neuropathy associated with a
disease or
disorder selected from the group consisting of acromegaly, ataxia
telangiectasia, Charcot-
Marie-Tooth disease, chronic obstructive pulmonary diseases, Fabry's disease,
Friedreich
ataxia, Guillain-Barre syndrome, hypoglycemia, IgG or IgA monoclonal
gammopathy
(non-malignant or associated with multiple myeloma or with osteosclerotic
myeloma),
lipoproteinemia, polycythemia vera, Refsum's syndrome, Reye's syndrome, and
Sjogren-
Larsson syndrome, a polyneuropathy associated with various drugs, with
hypoglycemia,
with infections such as HIV infection, or with cancer; epilepsy, amnesia,
anxiety,
hyperalgesia, psychosis, seizures, oxidative stress, opiate tolerance and
dependence, and
for the treatment of a psychosis or psychiatric disorder selected from the
group consisting
of an anxiety disorder, a mood disorder, schizophrenia or a schizophrenia-
related
disorder, drug use and dependence and withdrawal, and a memory loss or
cognitive
disorder.
In another embodiment, the use of the active agent in combination with stem
cell
therapy of the present invention is applied to injuries, diseases, disorders
or conditions
unrelated to the nervous system.
37
^,R,E4T.,` =*r,

CA 02588908 2010-12-07
In one preferred embodiment, the method is suitable for bone marrow-derived
stem cell transplantation for treatment of an injury, disease, disorder or
condition selected
from diabetes, failure of tissue repair, myocardial infarction, kidney
failure, liver
cirrhosis, muscular dystrophy, skin burn, leukemia, arthritis injury, or
osteoporosis injury.
The stem cells for use in the invention include, but are not limited to, adult
stem cells,
embryonic stem cells, umbilical cord blood stem cells, hematopoietic stem
cells,
peripheral blood stem cells, mesenchimal stem cells, multipotent stem cells,
neural stem
cells, neural progenitor cells, stromal stem cells, progenitor cells, or
precursors thereof,
and genetically-engineered stem cells, and any other stem cells that may be
found
suitable for the purpose of the present invention. Examples of such cells
include the CNS
neural stems cells disclosed in US 6,777,233 and US 6,680,198; the neural stem
cells and
hematopoietic cells disclosed in US 6,749,850 for administration with neural
stimulants;
and the stromal cells disclosed in US 6,653,134 for treatment of CNS diseases.
As used herein, the term "neural stem cell" is used to describe a single cell
derived
from tissue of the central nervous system, or the developing nervous system,
that can give
rise in vitro and/or in vivo to at least one of the following fundamental
neural lineages:
neurons (of multiple types), oligodendroglia and
38

CA 02588908 2012-11-02
astroglia as well as new neural stem cells with similar potential.
"Multipotent" or "pluripotent" neural stem cells are capable of giving rise to
all of
the above neural lineages as well as cells of equivalent developmental
potential.
In a more preferred embodiment, the neural stem cells are human neural stem
cells that can be isolated from both the developing and adult CNS, and can be
successfully grown in culture, are self-renewable, and can generate mature
neuronal
and ghat progeny. Embryonic human neural stern cells can be induced to
differentiate into specific neuronal phenotypes. Human neural stem cells
integrate
into the host environment after transplantation into the developing or adult
CNS.
Human neural stem cells transplanted into animal models of Parkinson's disease
and
spinal cord injury have induced functional recovery. However, there are still
problems with the engraftment of said cells and the present invention will
enhance
the successful engraftment, survival and further differentiation of the
implanted
cells. In a most preferred embodiment, the neural stem cells are autologous.
As used herein, the term "hematopoietic stem cells" refer to stem cells that
can give rise to cells of at least one of the major hematopoietic lineages in
addition
to producing daughter cells of equivalent potential. Certain hematopoietic
stem cells
are capable of giving rise to many other cell types including brain cells.
The stem cells, once isolated, are cultured by methods known in the art, for
example as described in US 5,958,767, US 5,270,191, US 5,753,506.
The treatment regimen according to the invention is carried out, in terms of
administration mode, timing of the administration, and dosage, depending on
the
type and severity of the injury, disease or disorder and the age and condition
of the
patient. The immunomodulator may be administered concomitanly with, before or
after the injection or implantation of the cells.
The administration of the cells may be carried out by various methods. In
certain embodiments, the cells are preferably administered directly into the
stroke
cavity, the spinal fluid, e.g., intraventricularly, intrathecally, or
intracistemally. The
stem cells can be formulated in a pharmaceutically acceptable liquid medium,
39

CA 02588908 2012-11-02
which can contain the Copolymer 1 or the T cells as well. Cells may also be
injected
into the region of the brain surrounding the areas of damage, and cells may be
given
systemically, given the ability of certain stem cells to migrate to the
appropriate
position in the brain.
In one preferred embodiment, the method of the invention comprises stem
cell therapy by administration of stem cells in combination with Copolymer 1.
In
one embodiment, the stem cells are injected/transplanted to the patient,
followed by
vaccination with Copolymer 1. In another embodiment, a combination of the stem

cells with the Copolymer 1 is injected/transplanted to the patient. In a
further
embodiment, the stem cells can be cultured in vitro (artificially) with the
Copolymer 1 and differentiated prior to transplantation
As used herein in the application, the terms "Cop 1", "Copolymer 1",
"glatiramer acetate" and "GA" are used interchangeably.
For the purpose of the present invention, " Copolymer 1 or a Copolymer 1-
related peptide or polypeptide" is intended to include any peptide or
polypeptide,
including a random copolymer that cross-reacts functionally with MBP and is
able
to compete with MBP on the MHC class II in the antigen presentation.
The composition for use in the invention may comprise as active agent a Cop
1 or a Cop 1-related peptide or polypeptide represented by a random copolymer
consisting of a suitable ratio of a positively charged amino acid such as
lysine or
arginine, in combination with a negatively charged amino acid (preferably in a

lesser quantity) such as glutamic acid or aspartic acid, optionally in
combination
with a non-charged neutral amino acid such as alanine or glycine, serving as a
filler,
and optionally with an amino acid adapted to confer on the copolymer
immunogenic properties, such as an aromatic amino acid like tyrosine or
tryptophan. Such compositions may include any of those copolymers disclosed in

WO 00/05250.
More specifically, the composition for use in the present invention comprises
at least one copolymer selected from the group consisting of random copolymers

CA 02588908 2012-11-02
comprising one amino acid selected from each of at least three of the
following
groups: (a) lysine and arginine; (b) glutamic acid and aspartic acid; (c)
alanine and
glycine; and (d) tyrosine and tryptophan.
The copolymers for use in the present invention can be composed of L- or D-
amino acids or mixtures thereof. As is known by those of skill in the art, L-
amino
acids occur in most natural proteins. However, D-amino acids are commercially
available and can be substituted for some or all of the amino acids used to
make the
copolymers used in the present invention. The present invention contemplates
the
use of copolymers containing both D- and L-amino acids, as well as copolymers
consisting essentially of either L- or D-amino acids.
In one embodiment of the invention, the copolymer contains four different
amino acids, each from a different one of the groups (a) to (d).
In a more preferred embodiment, the pharmaceutical composition or vaccine
of the invention comprises Copolymer 1, a mixture of random polypeptides
consisting essentially of the amino acids L-glutamic acid (E), L-alanine (A),
L-
tyrosine (Y) and L-lysine (K) in an approximate ratio of 1.5:4.8:1:3.6, having
a net
overall positive electrical charge and of a molecular weight from about 2 KDa
to
about 40 KDa.
In one preferred embodiment, the Cop 1 has average molecular weight of
about 2 KDa to about 20 KDa, more preferably of about 4,7 KDa to about 13 K
Da,
still more preferably of about 4 KDa to about 8.6 KDa, of about 5 KDa to 9
KDa, or
of about 6.25 KDa to 8.4 KDa. In another preferred embodiment, the Cop 1 has
average molecular weight of about 13 KDa to about 20 KDa, more preferably of
about 13,5 KDa to about 18 KDa, with an average of about 15 KDa to about 16
KD,
preferably of 16kDa. Other average molecular weights for Cop 1, lower than 40
KDa, are also encompassed by the present invention. Copolymer 1 of said
molecular weight ranges can be prepared by methods known in the art, for
example
by the processes described in U.S. Patent No. 5,800,808. The Copolymer 1 may
41

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
be a polypeptide comprising from about 15 to about 100, preferably from about
40
to about 80, amino acids in length.
In one preferred embodiment of the invention, the agent is Cop 1 in the form
of its acetate salt known under the generic name glatiramer acetate or its
trade name
Copaxone (a trademark of Teva Pharmaceutical Industries Ltd., Petach Tikva,
Israel).
The activity of Copolymer 1 for the composition disclosed herein is expected
to remain if one or more of the following substitutions is made: aspartic acid
for
glutamic acid, glycine for alanine, arginine for lysine, and tryptophan for
tyrosine.
In another embodiment of the invention, the Cop 1-related peptide or
polypeptide is a copolymer of three different amino acids each from a
different one
of three groups of the groups (a) to (d). These copolymers are herein referred
to as
terpolymers.
In one embodiment, the Cop 1-related peptide or polypeptide is a terpolymer
containing tyrosine, alanine, and lysine, hereinafter designated YAK, in which
the
average molar fraction of the amino acids can vary: tyrosine can be present in
a
mole fraction of about 0.05-0.250; alanine in a mole fraction of about 0.3 -
0.6; and
lysine in a mole fraction of about 0.1-0.5. More preferably, the molar ratios
of
tyrosine, alanine and lysine are about 0.10:0.54:0.35, respectively. It is
possible to
substitute arginine for lysine, glycine for alanine, and/or tryptophan for
tyrosine.
In another embodiment, the Cop 1-related peptide or polypeptide is a
terpolymer containing tyrosine, glutamic acid, and lysine, hereinafter
designated
YEK, in which the average molar fraction of the amino acids can vary: glutamic

acid can be present in a mole fraction of about 0.005 - 0.300, tyrosine can be
present
in a mole fraction of about 0.005-0.250, and lysine can be present in a mole
fraction
of about 0.3-0.7. More preferably, thé molar ratios of glutamic acid,
tyrosine, and
lysine are about 0.26:0.16:0.58, respectively. It is possible to substitute
aspartic acid
for glutamic acid, arginine for lysine, and/or tryptophan for tyrosine.
In another preferred embodiment, the Cop 1-related peptide or polypeptide is
a terpolymer containing lysine, glutamic acid, and alanine, hereinafter
designated
42

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
KEA, in which the average molar fraction of the amino acids can vary: glutamic

acid can be present in a mole fraction of about 0.005-0.300, alanine in a mole

fraction of about 0.005-0.600, and lysine can be present in a mole fraction of
about
0.2 - 0.7. More preferably, the molar ratios of glutamic acid, alanine and
lysine are
about 0.15:0.48:0.36, respectively. It is possible to substitute aspartic acid
for
glutamic acid, glycine for alanine, and/or arginine for lysine.
In a preferred embodiment, the Cop 1-related peptide or polypeptide is a
terpolymer containing tyrosine, glutamic acid, and alanine, hereinafter
designated
YEA, in which the average molar fraction of the amino acids can vary: tyrosine
can
be present in a mole fraction of about 0.005-0.250, glutamic acid in a mole
fraction
of about 0.005-0.300, and alanine in a mole fraction of about 0.005-0.800.
More
preferably, the molar ratios of glutamic acid, alanine, and tyrosine are about
0.21:
0.65:0.14, respectively. It is possible to substitute tryptophan for tyrosine,
aspartic
acid for glutamic acid, and/or glycine for alanine.
The average molecular weight of the terpolymers YAK, YEK, KEA and
YEA can vary between about 2 KDa to 40 KDa, preferably between about 3 KDa to
35 KDa, more preferably between about 5 KDa to 25 KDa.
Copolymer 1 and related peptides and polypeptides may be prepared by
methods known in the art, for example, under condensation conditions using the
desired molar ratio of amino acids in solution, or by solid phase synthetic
procedures. Condensation conditions include the proper temperature, pH, and
solvent conditions for condensing the carboxyl group of one amino acid with
the
amino group of another amino acid to form a peptide bond. Condensing agents,
for
example dicyclohexylcarbodiimide, can be used to facilitate the formation of
the
peptide bond. Blocking groups can be used to protect functional groups, such
as the
side chain moieties and some of the amino or carboxyl groups against undesired

side reactions.
For example, the copolymers can be prepared by the process disclosed in
U.S. Patent 3,849,550, wherein the N-carboxyanhydrides of tyrosine, alanine, y-

benzyl glutamate and N E-trifluoroacetyl-lysine are polymerized at ambient
43

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
temperatures (20 C-26 C) in anhydrous dioxane with diethylamine as an
initiator.
The y-carboxyl group of the glutamic acid can be deblocked by hydrogen bromide

in glacial acetic acid. The trifluoroacetyl groups are removed from lysine by
1M
piperidine. One of skill in the art readily understands that the process can
be
adjusted to make peptides and polypeptides containing the desired amino acids,
that
is, three of the four amino acids in Copolymer 1, by selectively eliminating
the
reactions that relate to any one of glutamic acid, alanine, tyrosine, or
lysine.
The molecular weight of the copolymers can be adjusted during polypeptide
synthesis or after the copolymers have been made. To adjust the molecular
weight
during polypeptide synthesis, the synthetic conditions or the amounts of amino
acids are adjusted so that synthesis stops when the polypeptide reaches the
approximate length that is desired. After synthesis, polypeptides with the
desired
molecular weight can be obtained by any available size selection procedure,
such as
chromatography of the polypeptides on a molecular weight sizing column or gel,
and collection of the molecular weight ranges desired. The copolymers can also
be
partially hydrolyzed to remove high molecular weight species, for example, by
acid
or enzymatic hydrolysis, and then purified to remove the acid or enzymes.
In one embodiment, the copolymers with a desired molecular weight may be
prepared by a process, which includes reacting a protected polypeptide with
hydrobromic acid to form a trifluoroacetyl-polypeptide having the desired
molecular weight profile. The reaction is performed for a time and at a
temperature
that is predetermined by one or more test reactions. During the test reaction,
the
time and temperature are varied and the molecular weight range of a given
batch of
test polypeptides is determined. The test conditions that provide the optimal
molecular weight range for that batch of polypeptides are used for the batch.
Thus,
a trifluoroacetyl-polypeptide having the desired molecular weight profile can
be
produced by a process, which includes reacting the protected polypeptide with
hydrobromic 'acid for a time and at a temperature predetermined by test
reaction.
The trifluoroacetyl-polypeptide with the desired molecular weight profile is
then
44

CA 02588908 2012-11-02
further treated with an aqueous piperidine solution to form a low toxicity
polypeptide having the desired molecular weight.
In a preferred embodiment, a test sample of protected polypeptide from a
given batch is reacted with hydrobromic acid for about 10-50 hours at a
temperature
of about 20-28 C. The best conditions for that batch are determined by running
several test reactions. For example, in one embodiment, the protected
polypeptide is
reacted with hydrobromic acid for about 17 hours at a temperature of about 26
C.
As binding motifs of Cop I to MS-associated HLA-DR molecules are known
(Fridkis-Hareli et al, 1999), polypeptides derived from Cop 1 having a defined

sequence can readily be prepared and tested for binding to the peptide binding

groove of the HLA-DR molecules as described in the Fridkis-Hareli et al (1999)

publication. Examples of such peptides are those disclosed in WO 00/05249 and
WO 00/05250, and include the peptides of SEQ ID NOs. 1-32 hereinbelow.
SEQ ID NO. Peptide Sequence
1 AAAYAAAAAAKAAAA
2 AEKYAAAAAAKAAAA
3 AK EYAAAAAAKAAAA
4 AKKYAAAAAAKAAAA
5 AEAYAAAAAAKAAAA
6 KEAYAAAAAAKAAAA
7 A EEYAAAAAAKAAAA
8 AAEYAAAAAAKAAAA
9 EKAYAAAAAAKAAAA
10 AAKYEAAAAAKAAAA
11 AAKYAEAAAAKAAAA
12 EAAYAAAAAAKAAAA
13 EKKYAAAAAAKAAAA
14 EAKYAAAAAAKAAAA
15 AEKYAAAAAAAAAAA
16 AKEYAAAAAAAAAAA
17 AKKYEAAAAAAAAAA
18 AKKYAEAAAAAAAAA
19 AEAYKAAAAAAAAAA
20 KEAYAAAAAAAAAAA

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
21 AEEYKAAAAAAAAAA
22 AAEYKAAAAAAAAAA
23 EKAYAAAAAAAAAAA
24 AAKYEAAAAAAAAAA
25 AAKYAEAAAAAAAAA
26 EKKYAAAAAAAAAAA
27 EAKYAAAAAAAAAAA
28 AEYAKAAAAAAAAAA
29 AEKAYAAAAAAAAAA
30 EKYAAAAAAAAAAAA
31 AYKAEAAAAAAAAAA
32 AKYAEAAAAAAAAAA
Such peptides and other similar peptides derived from Cop 1 would be
expected to have similar activity as Cop 1. Such peptides, and other similar
peptides, are also considered to be within the definition of Cop 1-related
peptides or
polypeptides and their use is considered to be part of the present invention.
The definition of "Cop 1-related peptide or polypeptide" according to the
invention is meant to encompass other synthetic amino acid copolymers such as
the
random four-amino acid copolymers described by Fridkis-Hareli et al., 2002 (as

candidates for treatment of multiple sclerosis), namely copolymers (14-, 35-
and 50-
mers) containing the amino acids phenylalanine, glutamic acid, alanine and
lysine
(poly FEAK), or tyrosine, phenylalanine, alanine and lysine (poly YFAK), and
any
other similar copolymer to be discovered that can be considered a universal
antigen
similar to Cop 1.
The dosage of Cop 1 to be administered will be determined by the physician
according to the age of the patient and stage of the disease and may be chosen
from
a range of 1-80 mg, preferably 20 mg, although any other suitable dosage is
encompassed by the invention. The treatment should be preferably carried out
by
administration of repeated doses at suitable time intervals, according to the
neurodegenerative disease to be treated, the age and condition of the patient.
In one
embodiment, Cop 1 may be administered daily. In another embodiment, the
administration may be made according to a regimen suitable for immunization,
for
example, at least once a month or at least once every 2 or 3 months, or less
46

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
frequently, but any other suitable interval between the immunizations is
envisaged
by the invention according to the condition of the patient.
Pharmaceutical compositions for use in accordance with the present
invention may be formulated in conventional manner using one or more
physiologically acceptable carriers or excipients.
The carrier(s) must be
"acceptable" in the sense of being compatible with the other ingredients of
the
composition and not deleterious to the recipient thereof.
Methods of administration include, but are not limited to, parenteral, e.g.,
intravenous, intraperitoneal, intramuscular, subcutaneous, mucosal (e.g.,
oral,
intranasal, buccal, vaginal, rectal, intraocular), intrathecal, topical and
intradermal
routes, with or without adjuvant. Administration can be systemic or local.
The invention will now be illustrated by the following non-limiting
examples.
EXAMPLE 1
Microglia induce neural cell renewal - Microglia activated by IL-4 or IFNI
differentially induce neurogenesis and oligodendrogenesis from adult stem/
progenitor cells
Materials and Methods
(i) Animals. Tneonatal (PO-P1) C57B1/6J mice were supplied by the Animal
Breeding Center of the Weizmann Institute of Science (Rehovot, Israel). All
animals were handled according to the regulations formulated by the Weizmann
Institute's Animal Care and Use Committee.
(it) Reagents. Lipopolysaccharide (LPS) (containing (1% contaminating
proteins) was obtained from Escherichia coli 0127:B8 (Sigma-Aldrich, St.
Louis,
MO). Recombinant mouse tumor necrosis factor (TNF)-a and insulin-like growth
factor (IGF)-I (both containing endotoxin at a concentration below 1 EU per
pig of
cytokine), recombinant rat and mouse interferon (IFN)-y and interleukin (IL)-4

(both containing endotoxin at a concentration below 0.1 ng per pig of
cytokine),
47

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
goat anti-mouse neutralizing anti-TNF-a antibodies (a"TNF-a; containing
endotoxin
at a concentration below 0.001 EU per pg of Ab), and goat anti-mouse
neutralizing
anti-IGF-I (aIGF-I; containing endotoxin at a concentration below 0.1 EU per
lig of
Ab) were obtained from R&D Systems (Minneapolis, MN).
(iii) Neural progenitor cell (NPC) culture. Coronal sections (2 mm thick) of
tissue containing the subventricular zone of the lateral ventricle were
obtained from
the brains of adult C57B16/J mice. The tissue was minced and then incubated
for
digestion at 37 C, 5% CO2 for 45 min in Earle's balanced salt solution
containing
0.94 mg/ml papain (Worthington, Lakewood, NJ) and 0.18 mg/ml of L-cysteine and
EDTA. After centrifugation at 110 x g for 15 min at room temperature, the
tissue
was mechanically dissociated by pipette trituration. Cells obtained from
single-cell
suspensions were plated (3500 cells/cm2) in 75-cm2 Falcon tissue-culture
flasks
(BD Biosciences, Franklin Lakes, NJ), in NPC-culturing medium [Dulbecco's
modified Eagles's medium (DMEM)/F12 medium (Gibco/Invitrogen, Carlsbad,
CA) containing 2 mM L-glutamine, 0.6% glucose, 9.6 ig/ml putrescine, 6.3 ng/ml
progesterone, 5.2 ng/ml sodium selenite, 0.02 mg/ml insulin, 0.1 mg/ml
transferrin,
2 jig/m1 heparin (all from Sigma-Aldrich, Rehovot, Israel), fibroblast growth
factor-
2 (human recombinant, 20 ng/ml), and epidermal growth factor (human
recombinant, 20 ng/ml; both from Peprotech, Rocky Hill, NJ)]. Spheres were
passaged every 4-6 days and replated as single cells. Green fluorescent
protein
(GFP)-expressing neural progenitor cells (NPCs) were obtained as previously
described (Pluchino et al., 2003).
(iv) Primary microglial culture. Brains from neonatal (P0¨P1) C57B1/6J
mice were stripped of their meninges and minced with scissors under a
dissecting
microscope (Zeiss, Stemi DV4, Germany) in Leibovitz-15 medium (Biological
Industries, Beit Ha-Emek, Israel). After trypsinization (0.5% trypsin, 10 min,

37 C/5% CO2), the tissue was triturated. The cell suspension was washed in
culture
medium for glial cells [DMEM supplemented with 10% fetal calf serum (FCS;
Sigma-Aldrich, Rehovot), L-glutamine (1 mM), sodium pyruvate (1 mM),
penicillin
(100 U/ml), and streptomycin (100 mg/ml)] and cultured at 37 C/5% CO2 in 75-
cm2
48

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Falcon tissue-culture flasks (BD Biosciences) coated with poly-D-lysine (PDL)
(10
mg/ml; Sigma-Aldrich, Rehovot) in borate buffer (2.37 g borax and 1.55 g boric

acid dissolved in 500 ml sterile water, pH 8.4) for 1 h, then rinsed
thoroughly with
sterile, glass-distilled water. Half of the medium was changed after 6 h in
culture
and every 2nd day thereafter, starting on day 2, for a total culture time of
10-14
days. Microglia were shaken off the primary mixed brain glial cell cultures
(150
rpm, 37 C, 6 h) with maximum yields between days 10 and 14, seeded (105
cells/m1) onto PDL-pretreated 24-well plates (1 ml/well; Corning, Corning,
NY),
and grown in culture medium for microglia [RPMI-1640 medium (Sigma-Aldrich,
Rehovot) supplemented with 10% FCS, L-glutamine (1 mM), sodium pyruvate (1
mM), 13-mercaptoethano1 (50 mM), penicillin (100 U/ml), and streptomycin (100
mg/m1)]. The cells were allowed to adhere to the surface of a PDL-coated
culture
flask (1 h, 37 C/5% CO2), and non-adherent cells were rinsed off.
(v) Co-culturing of neural progenitor cells (NPCs) and mouse microglia.
Microglia were treated for 24 h with cytokines (IFN-y, 20 ng/ml; IL-4, 10
ng/ml) or
LPS (100 ng/ml). Cultures of treated or untreated microglia were washed twice
with
fresh NPC-differentiation medium (same as the culture medium for NPCs but
without growth factors and with 2.5% FCS) to remove all traces of the tested
reagents, then incubated on ice for 15 min, and shaken at 350 rpm for 20 min
at
room temperature. Microglia were removed from the flasks and immediately co-
cultured (5 x 104 cells/well) with NPCs (5 x 104 cells/well) for 5 or 10 days
on
cover slips coated with Matrigel (BD Biosciences) in 24-well plates, in the
presence
of NPC differentiation medium, with or without insulin. The cultures were then

fixed with 2.5% paraformaldehyde in PBS for 30 min at room temperature and
stained for neuronal and glial markers. Cell proliferation rates and cell
survival in
vitro were determined by staining with 5-bromo-2'-deoxyuridine (BrdU, 2.5
vt,M;
Sigma-Aldrich, St. Louis). For quantification of live and dead cells, live
cultures
were stained with 1 Ag/m1 propidium iodide (Molecular Probes, Invitrogen,
Carlsbad, CA) and 1 1.1.g/m1 Hoechst 33342 (Sigma-Aldrich, St. Louis), and
cells
49

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
were counted using Image-Pro (Media Cybernetics, Silver Spring, MD), as
described (Hsieh et al., 2004)
(vi) Immunocytochemistry. Cover slips from co-cultures of NPCs and mouse
microglia were washed with PBS, fixed as described above, treated with a
permeabilization/blocking solution containing 10% FCS, 2% bovine serum
albumin, 1% glycine, and 0.1% Triton X-100 (Sigma-Aldrich, Rehovot) and
stained
with a combination of the mouse or rabbit anti-tubulin 13-III-isoform C-
terminus
antibodies (P-III-tubulin; 1:500), rabbit anti-NG2 chondroitin sulfate
proteoglycan
(NG2; 1:500), mouse anti-RIP (RIP; 1:2000), mouse anti-galactocerebroside
(GalC;
1:250), mouse anti-glutamic acid decarboxylase 67 (GAD; 1:1000), mouse anti-
nestin (Nestin; 1:1000), rat anti-myelin basic protein (MBP; 1:300) (all from
Chemicon, Temecula, CA), goat anti-double cortin (DCX; 1:400, Santa Cruz
Biotechnology, Santa Cruz, CA) and mouse anti-glial fibrillary acidic protein
(GFAP; 1:100, Sigma-Aldrich, St. Louis). For labeling of microglia we used
either
rat andi-CD1 lb (MAC1; 1:50, BD-Pharmingen, NJ) or FITC-conjugated
Bandeiraea sinplicfolia isolectin B4 (IB4; 1:50, Sigma-Aldrich, Rehovot).
Expression of IGF-1 was detected by goat anti-IGF-1 (1:20, R&D Systems).
(vii) RNA purification, cDNA synthesis, and reverse-transcription PCR
analysis. Cells were lysed with TRI reagent (MRC, Cincinnati, OH), and total
cellular RNA was purified from lysates using the RNeasy kit (Qiagen, Hilden,
Germany) according to the manufacturer's instructions. Residual genomic DNA
was removed during the purification process by incubation with RNase-free
DNase
(Qiagen). RNA was stored in RNase-free water (Qiagen) at ¨80 C. RNA (1 lig)
was
converted to cDNA using SuperScript II (Promega, Madison, WI), as recommended
by the manufacturer. The cDNA mixture was diluted 1:5 with PCR-grade water.
We assayed the expression of specific mRNAs using semi-quantitative
reverse transcription PCR (RT¨PCR) with selected gene-specific primer pairs,
using OLIGO v6.4 (Molecular Biology Insights, Cascade, CO).
The primers used were:
TNF-a, sense 5'-AGGAGGCGCTCCCCAAAAAGATGGG-3' (SEQ ID NO:33),

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
antisense 5'-GTACATGGGCTCATACCAGGGCTTG-3' (target size, 551 bp)
(SEQ ID NO:34);
IGF-I, sense 5'-CAGGCTCCTAGCATACCTGC-3' (SEQ ID NO:35),
antisense 5'-GCTGGTAAAGGTGAGCAAGC-3' (target size, 244 bp) (SEQ ID
NO:36); and
[3-actin, sense 5'-TTGTAACCAACTGGGACGATATGG-3' (SEQ ID NO:37),
antisense 5'-GATCTTGATCTTCATGGTGCTAGG-3' (target size, 764 bp) (SEQ
ID NO:38).
The RT¨PCR reactions were carried out using 1 lag of cDNA, 35 nmol of
each primer, and ReadyMix PCR Master Mix (ABgene, Epsom, UK) in 30- .1
reactions. PCR reactions were carried out in an Eppendorf PCR system with
cycles
(usually 25-30) of 95 C for 30 s, 60 C for 1 min, 72 C for 1 min, and 72 C for
5
min, and then kept at 4 C. As an internal standard for the amount of cDNA
synthesized, we used 13-actin mRNA. PCR products were subjected to agarose gel
analysis and visualized by ethidium bromide staining. Signals were quantified
using
a Gel-Pro analyzer 3.1 (Media Cybernetics). In all cases one product was
observed
with each primer set, and the observed product had an amplicon size that
matched
the size predicted from published cDNA sequences.
(viii) Quantification. For microscopic analysis we used a Zeiss LSM 510
confocal laser scanning microscope (40x magnification). For experiments in
vitro
we scanned fields of 0.053 mm2 (n = 8-16 from at least two different
coverslips) for
each experimental group. For each marker, 500-1000 cells were sampled. Cells
co-
expressing GFP and p-III-tubulin, NG2, RIP, GalC, and GFAP were counted.
(Lx) Statistical analysis. The results were analyzed by the Tukey¨Kramer
multiple comparisons test (ANOVA) and are expressed as means SD (unless
differently indicated).
51

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Example 1(1). Effect of microglia on neurogenesis in vitro - Microglia
pretreated with IL-4 or IFNI induce and support neuronal differentiation
from neural progenitor cells (NPCs) in vitro.
Adaptive immunity, in the form of a well-controlled Thl or a Th2 response
to a CNS insult, induces microglia (MG) to adopt a phenotype that facilitates
neuronal protection and neuronal tissue repair (Butovsky et al., 2001). Here
we
examined the ability of adaptive immunity, via activation of microglia, to
induce or
support the differentiation of NPCs. Neurogenesis is reportedly blocked by the

inflammation caused by microglia activated with endotoxin (such as
lipopolysaccharide, LPS) (Ekdahl et al., 2003). We therefore compared the
effects
on NPCs of microglia exposed to LPS (MG(Lps)) with the effects of microglia
exposed to the low levels of characteristic Thl (pro-inflammatory) and Th2
(anti-
inflammatory) cytokines, IFN-Y (MG(IFN and IL-4 (MG447)),
-7)) respectively,
shown
herein to be supportive of neural survival. We used NPCs expressing green
fluorescent protein (GFP) to verify that any neural cell differentiation seen
in the
culture was derived from the NPCs rather than from contamination of the
primary
microglial culture.
Microglia were grown in their optimal growth medium (Zielasek et al., 1992)
and were then treated for 24 h with IL-4, IFN-y (low level), or LPS. Residues
of the
growth medium and the cytokines were washed off, and each of the treated
microglial preparations, as well as a preparation of untreated microglia
(MG()), was
freshly co-cultured with dissociated NPC spheres in the presence of
differentiation
medium. We examined the effects of both IFN-y-activated and IL-4-activated
microglia. After 5 days in culture, GFP+ cells that expressed the neuronal
marker 13-
III tubulin were identified as neurons.
Since we recently showed that IL-4-activated. microglia (MG(11,-4)) produce a
high level of IGF-1 (Butovsky et al., 2005), and because IGF-1 is reportedly a
key
factor in neural cell renewal (O'Kusky et al., 2000), we envisioned a
situation in
which IGF-1 might be one of the factors in the effect of IL-4-activated
microglia.
Therefore, the following experiments were carried out both in insulin-free (to
allow
52

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
detection, if exists, of the effect of insulin-related factors secreted by the
activated
microglia) and in insulin-containing differentiation media.
Quantitative analysis revealed that neurogenesis, in the absence of insulin,
was only minimally supported by MG(I) and was impaired by MG(Lps), but was
almost 3-fold higher in NPCs co-cultured with MG(IL.4) than in controls (Fig.
1A).
In the presence of insulin the picture was somewhat different: MG(IF) were
significantly more effective than MG(_) in inducing neurogenesis, whereas the
inductive effect of MG(11_,_4) and the blocking effect of MG(LPs) on
neurogenesis were
similar to their effects in the absence of insulin (compare Fig. 1B). In the
absence of
microglia, addition of insulin (0.02 mg/ml) did not increase the numbers of
GFP-1713-
III-tubu1in+ cells in NPC cultures (Fig. 1A).
In co-cultures of NPCs with MGH, however, addition of insulin increased
the percentage of GFP+/P-III-tubulin+ cells (Fig. 1B) relative to their
percentage in
such co-cultures without insulin (Fig. 1B) or in control (microglia-free)
cultures in
insulin-containing medium (Fig. 1B). In the presence of insulin, the number of
neurons in NPCs co-cultured with MG(I) (Fig. 1B) was greater than in NPCs co-
cultured with MG(_) (Fig. 1B), and even greater if the NPCs were co-cultured
with
MG(IFNI) containing neutralizing anti-TNF-a antibodies (aTNF-a) (Fig. 1B).To
verify that the observed beneficial effect of aTNF-a in the MG(E,T_y) co-
cultures
(Fig. 1B) was due to neutralization of the adverse effect of TNF-a on
neurogenesis,
we added recombinant mouse TNF-a (rTNF-a) to NPCs freshly co-cultured with
MG(FN..y). Fig. 1C shows that in the presence of rTNF-a the numbers of GFP+/13-
III-
tubulin+ cells were similar to those in control (untreated) NPC cultures.
Morphological differences were observed between the newly differentiating
neurons in NPCs co-cultured with MG(IFN..y) and those generated in co-cultures
with
MG-(.4) (Fig. 1D). Co-expression of GFP with p-III-tubulin is shown in Fig.
1E.
The newly differentiating neurons were positively labeled for GAD67 (glutamic
acid decarboxylase 67), an enzyme responsible for the synthesis of GABA, the
major inhibitory transmitter in higher brain regions, and were also found to
be co-
53

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
labeled with GFP (13-III-tubu1in+/GFP+/GAD+) (Fig. 1F). In another set of
experiments we prepared cultures similar to those described above and stained
them
with doublecortin (DCX; Fig. 2), a marker of early differentiation of the
neuronal
lineage. This staining revealed a similar effect of the various microglial
preparations
to that seen with staining for P-III-tubulin. Striking differences in the
morphology
of newly differentiating neurons were seen between NPCs co-cultured with MG(1i-
4)
and those co-cultured with MG(I) (Fig. 2A); the former showed significant
branching, whereas in the latter the neurons were polarized and had long
processes
(Fig. 2A). These differences suggested that the mechanisms activated in
microglia
by the two cytokines are not identical. Co-expression of GFP with DCX is shown
in
Fig. 2B. In cultures stained for both DCX and P-III-tubulin, these two
neuronal
markers were found to be co-localized (Fig. 2C). In all of the above
experiments
microglial viability, assayed by propidium iodide staining of live cells
(Hsieh et al.,
2004), was unaffected by the co-culturing conditions. Quantitative analysis of
GFP/DCX-stained cells, shown in Fig. 2D, yielded similar results to those
obtained when P-III-tubulin was used as the neuronal marker (Figs. 1A, 1B).
Example 1(2). Effect of microglia on oligodendrogenesis in vitro -
Differentiation of NPCs into oligodendrocytes is induced by co-culturing with
IL-4 pretreated microglia (MG(w-4))
Next we examined whether, under the same experimental conditions,
microglia would also induce NPCs to differentiate into oligodendrocytes. Under

high magnification, we were able to detect newly formed oligodendrocytes. In
attempting to detect possible differentiation of NPCs to oligodendrocytes, we
first
looked for GFP-labeled cells co-expressing oligodendrocyte progenitor marker
NG2. Quantitative analysis confirmed that both MG(IL-4) and (to a lesser
extent)
MG(fl) induced differentiation of NG2+ cells from co-cultured NPCs (Figs. 3A,
3B). In both MG(_) and MG(I) co-cultured with NPCs, significantly fewer NG2+-
expressing cells were seen in the absence of insulin (Fig. 3A) than in its
presence
(Fig. 3B). Unlike in the case of neurogenesis (Fig. 1), MG(IFN.7)¨even in the
54

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
presence of insulin¨was significantly less effective than MG(JL..4) in
inducing the
appearance of newly differentiating oligodendrocytes (I\TG2+). A significant
proportion of the NG2+ cells were also labeled for RIP [a monoclonal antibody
that
specifically labels the cytoplasm of the cell body and processes of premature
and
mature oligodendrocytes at the pre-ensheathing stage (Hsieh et al., 2004)] in
both
the MG(J) and the MG(IL-4) co-cultures (Figs. 3A, 3B). In the absence of
insulin,
almost no GFP+/NG2+ cells were seen in control medium containing no microglia
(Fig. 3A). A few GFP+/NG2+ cells were seen in co-cultures of NPCs with MG(_)
(Fig. 3A), but none in co-cultures with MG(LPs) (Fig. 3A). A dramatic increase
in the
numbers of these cells was seen in co-cultures with MG(11,-4) (Figs. 3A, 3C).
Addition of insulin to the NPC cultures did not affect the incidence of NG2+
cells in the absence of microglia (control; Fig. 3B); it did, however, cause
an
increase in the numbers of NG2+ cells in NPCs co-cultured with MG(_) (Fig.
3B).
Moreover, in the presence of insulin the blocking effect of MG(LPs) on newly
differentiating NG2+ cells was not altered (Fig. 3B), whereas the numbers of
NG2+
cells in co-cultures with MG(IF) were increased (Figs. 3B, 3C). In each of the
co-
cultures, all NG2+ cells were also found to be labeled with GFP (Fig. 3D). An
interesting observation was the close spatial association between the
microglia and
the newly differentiating oligodendrocytes (Fig. 3E).
In light of the observed early differences between the effects of MG(IL..4)
and
MG(I) on both neurogenesis and oligodendrogenesis, we examined NPCs co-
cultured with the cytokine-activated microglia after 10 days in co-culture. As
on day
5, few NG2+ cells were seen in the absence of microglia (Fig. 4A).
Quantitative
analysis of these cultures disclosed striking differences: while both of the
cytokine-
activated microglial preparations induced differentiation to both
oligodendrocytes
(NG2+, RIP , Ga1C
++) and neurons (13-III-tubuli+ n), MG(IL_4) showed a positive bias
towards mature oligodendrocytes and MG(JFN.y) towards mature neurons. Analysis

of the incidence of astrocytes (GFAP+ cells) in these cultures (after 10 days
of co-
culturing) disclosed no significant differences between co-cultures of NPCs
with

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
MG(1L-4) and with MG(IFN.y); in both, however, GFAP+ cells were more numerous
than in NPC cultures without microglia (Fig. 4A). The results suggested that
these
two types of cytokine-activated microglia affect the three neural cell
lineages, and
that they have different effects on the neuronal and oligodendrocyte lineages
but not
on the astrocytic lineage (Fig. 4A).
In the presence of MG(1L-4), the GFP+/NG2+ cells were more branched at 10
days (Fig. 4B) than at 5 days (Fig. 3C). The branching cells appeared to be
forming
contacts with cells that looked like neurons (Fig. 4B). Staining of these
cultures for
galactocerebroside (GalC), a marker of mature oligodendrocytes, and for the
neuronal marker 13-III-tubulin, verified that the contact-forming cells were
newly
formed oligodendrocytes and neurons (Fig. 4C). Analysis of the same cultures
for
DCX and RIP (Fig. 4D) revealed that none of the newly differentiating cells
expressed both of these markers together. Moreover, there was no overlapping
in
expression of the astrocyte marker glial fibrillary acid protein (GFAP) and
NG2
(Fig. 4E) or of GFAP and DCX (Fig. 4F). Analysis of neurite length induced by
the
cytokine-activated microglia (after 10 days of co-culturing) revealed that
neurites of
the newly formed neurons in NPCs co-cultured with MG(i7) were significantly
longer than in NPCs co-cultured with MG(1L4) or in NPCs alone, with no
significant
differences between neurite lengths in the latter two (Fig. 4G).
Interestingly, there
were no significant differences between the absolute numbers of GFP+ cells
counted
in these three groups (NPCs alone: 90.2 32.0; co-cultured with MG(JFN..y):
70.5 23.0; co-cultured with MG(4): 66.1 10.4). This raises a question: do the
activated microglia, besides affecting differentiation, also affect NPC
proliferation
and/or survival?
Table 1 records the proliferation of NPCs co-cultured with non-activated,
IL-4-activated, or IFN-y-activated microglia. Cultures of untreated NPCs
(control)
or NPCs co-cultured with MG(_) or MGRA or MG(I7) or MG(Lps), with or without
insulin, were analyzed for proliferation and cell death 24, 48, or 72 h after
plating.
For the proliferation assay, a pulse of BrdU was applied 12 h before each time
point. Numbers of BrdU+ cells are expressed as percentages of GFP+ cells (mean
56

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
SEM from three independent experiments in duplicate) and analyzed by ANOVA.
Cell death with and without insulin was determined by live staining with 1
g/m1
propidium iodide and 1 tig/m1 Hoechst 33342 (mean SEM from two independent =

experiments in duplicate; * P < 0.05; * * * P <0.001; ANOVA).
As shown in Table 1, comparisons of proliferation at 24 h and 48 h of culture
revealed no differences. After 72 h a slight but non-significant difference
was seen
between NPCs alone and NPCs co-cultured with MG(_) or MG(IFN1), possibly
because of decreased proliferation in the culture of NPCs alone rather than
any
increase in the co-cultures. In the absence of insulin there were no
significant
differences at any time in culture between NPCs alone and NPCs co-cultured
with
MG(_) or with MG(1L_4). A reduction in proliferation was observed in NPCs co-
cultured with MGLps, with or without insulin. After 5 days, no proliferation
was
detectable in any of the co-cultures (data not shown). To identify dead or
dying cells
we stained live cultures with 1 g/ml propidium iodide, which stains dead
cells, and
1 g/ml Hoechst 33342, which stains both live and dead cells (Hsieh et al.,
2004).
Significant cell death was observed in NPCs co-cultured with MG(Lps), both in
the
absence and in the presence of insulin, whereas in NPCs cultured alone or with

MG(I) Or MG(11,-4) the percentage of cell death was low and did not differ
significantly from that seen in cultures of NPCs alone (Table 1). These
results
suggested that the primary effect of the cytokine-activated microglia on the
fate of
NPCs in vitro occurs via a mechanism that is instructive rather than
selective.
57

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Table 1. Proliferation and survival of neural progenitor cells (NPCs) in co-
cultures with microglia
% BrdU+ cells out of GFP+ cells % Pr cells out of GFP+ cells
+Insulin +Insulin
Treatment Treatment
24h 48h 72h 24h 48h 72h
Control 5.8 0.6 5.4 1.5 1.410.9 Control 3.3 0.9 1.710.4 1.7 0.7
MG() 6.2 0.5 6.9 1.9 5.6 1.4 MG() 4.010.5 2.8 0.7 2.8 0.9
MG(IFN_y) 5.8 1.5 7.1 2.5 5.611.1 MG(IFN1,) 5.9 0.6 4.2 1.5 3.6 2.1
_
MG(LPs) 3.1 0.8 1.710.4 1.110.1 MG(LPs) 14.0 0.1*** 7.311.9 4.1 0.2
-Insulin -Insulin
Treatment Treatment
24h 48h 72h 24h 48h 72h
Control 4.1 0.6 3.210.5 1.2 0.7 Control 3.7 0.2 2.3+0.8 2.2 1.1.
MG() 6.5 1.5 3.5 0.7 3.3 2.5 MG() 5.2 3.0 3.7+0.3 3.0 1.0
_
MG(IFN-4) 6.2 1.1 5.411.9 3.3 2.2 MG(IFN-4) 3.7 2.0 2.9+0.4 2.5 0.3
_
MG(Lps) 2.1 0.2 1.7 0.5 1.1 0.6 MG(LPs) 15.8 1.9* 7.0+5.0 4.8 0.8
Proliferation and survival of neural progenitor cells (NPCs) in co-cultures
with microglia.
Cultures of untreated NPCs (control) or NPCs co-cultured with MG(_) or
MG(1L_4) or
MG(IFN_y) or MG(Lps), with or without insulin, were analyzed for proliferation
and cell death
24, 48, or 72 h after plating. For the proliferation assay a pulse of BrdU was
applied 12 h
before each time point. Numbers of BrdU+ cells are expressed as percentages of
GFP+ cells
(mean SEM from three independent experiments in duplicate) and analyzed by
ANOVA.
Cell death with and without insulin was determined by live staining with 1
1...tg/m1
propidium iodide and 1 ptg/m1 Hoechst 33342 (mean SEM from two independent
experiments in duplicate; * P < 0.05; *** P ( 0.001; ANOVA).
Example 1(3). Possible mechanism of oligodendrogenesis induction by IL-4-
and IFN-y-activated microglia
Insulin-like growth factor (IGF)-I is reportedly a key factor in neurogenesis
and oligodendrogenesis (Carson et al., 1993; Aberg et al., 2000; O'Kusky et
al.,
2000; Hsieh et al., 2004). To determine whether the beneficial effect of the
cytokine-activated microglia on the differentiation of NPCs is mediated, at
least in
part, by the ability of the microglia to produce IGF-I, we added neutralizing
antibodies specific to IGF-I (dIGF-I) to the NPCs co-cultured with activated
microglia. aIGF-I blocked the MG(1L4)-induced effect on oligodendrogenesis
(Fig.
58

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
5A), indicating that the effect of IL-4-activated microglia on
oligodendrogenesis is
dependent on IGF-I. Direct addition of recombinant IGF-I (rIGF-I; 500 ng/ml)
to
NPCs resulted in their significant differentiation to NG2-expressing cells
(Fig. 58).
Such differentiation, however, was less extensive than that observed in NPCs
co-
cultured with MG(1L4) (Fig. 5A), suggesting that the MG(IL.4) effect is
mediated
through additional (possibly soluble) factors, or by cell¨cell interaction, or
both.
alIGF-I had no effect on oligodendrogenesis induced by MG(IFNi,) (data not
shown).
We also examined the effect of dIGF-I on MG(1L4)-induced neurogenesis by
assessing 13-III-tubu1in expression. The percentage of GFP+/13-III-tubu1in+
cells was
21.9 2.9% in NPCs co-cultured with MG(1L4) and 19.7 4.5%, (P = 0.3) when
aIGF-I was added to those co-cultures. These results suggested that MG(IL4)
produces additional potent neurogenic factors besides IGF-I.
In light of the observed beneficial effect of aINF-a on the outcome of
MG(IFN.y)-induced neurogenesis (Fig. 1), we examined whether neutralization of
TNF-a would promote MG(IFN.y)-induced oligodendrogenesis as well.
Oligodendrogenesis was indeed enhanced by 5INF-a in NPCs co-cultured with
MG(JD-N.y) (Fig. 5C). The implied negative effect of TNF-a was substantiated
by
direct addition of TNF-a to NPCs co-cultured with MG(IFN_y) (Fig. 5D).
Comparative RT¨PCR analyses of microglial mRNA disclosed that in the
absence of activation the microglia produced both IGF-I and low levels of TNF-
a.
Analysis of TNF-a and IGF-I production as a function of time revealed that IFN-
y,
unlike IL-4, caused a transient increase in TNF-a production and down-
regulation
of IGF-I (Figs. 6A, 6B). At the protein level, quantitative immunocytochemical

analysis also disclosed up-regulation of the expression of IGF-I by MG(JL_4).
LPS
completely blocked the production of IGF-I (Fig. 6C).
Discussion
The results of this study strongly suggest that certain specifically activated

microglia can induce neural cell renewal in the adult CNS. The findings showed
59

CA 02588908 2007-05-29
WO 2006/057003
PCT/1L2005/001275
that microglia can determine the fate of differentiating adult NPCs. Both
neurogenesis and oligodendrogenesis were induced in NPCs co-cultured with
MG(IL_4),and MG(IFN_y), whereas both were blocked by MG(Lps), in line with
reports
that inflammation associated with LPS blocks adult neurogenesis (Ekdahl et
al.,
2003; Monje et al., 2003). NPCs co-cultured with MG(1L4) showed a bias towards
oligodendrogenesis, whereas NPCs co-cultured with MG(I) were biased towards
neurogenesis.
EXAMPLE 2
Synergy between T cells and adult neural progenitor cells promotes
functional recovery from spinal cord injury
Recovery from spinal cord injury evidently necessitates a local immune
response that is amenable to well-controlled boosting by immunization with T
lymphocytes recognizing myelin-associated antigens at the injury site. The
relevant
T cells can activate local microglia to express a phenotype supportive of
neuronal
survival and renewal. We show that recovery of mice from spinal contusion is
synergistically promoted by T-cell-based vaccination with a myelin-derived
peptide
and injection of adult neural stem/progenitor cells (aNPCs) into the
cerebrospinal
fluid. Significantly more aNPCs targeted the lesion site in vaccinated than in
nonvaccinated mice. Synergistic interaction between aNPCs and T cells in vitro
was
critically dependent on T-cell specificity and phenotype. The results suggest
that
controlled immune activity underlies efficient regulation of the stem-cell
niche, and
that stem-cell therapy necessitates autologous or histocompatibility-matched
donors
instead of the immunosuppressive anti-rejection drugs that would eliminate any
beneficial effect of immune cells on spinal cord repair.
Materials and Methods
(x) Animals. Inbred adult wild-type C57B1/6J mice were supplied by the
Animal Breeding Center of The Weizmann Institute of Science. All animals were

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
handled according to the regulations formulated by the Institutional Animal
Care
and Use Committee (IACUC).
(xi) Antigens. The following peptides were synthesized by the Synthesis
Unit at the Weizmann Institute (Rehovot, Israel) : MOG, residues 35-55
MEVGWYRSPFSRVVHLYRNGK (SEQ ID NO:39) and an altered MOG peptide
(45D) MEVGWYRSPFDRVVHLYRNGK (SEQ ID NO:40), a peptide analog of
MOG 35-55 containing a serine to aspartic acid substitution as shown. OVA was
purchased from Sigma.
(xii) Immunization. Adult mice were immunized with MOG, 45D, or OVA
(all 100 ig), each emulsified in an equal volume of CFA (Difco, Detroit, MI)
containing Mycobacterium tuberculosis (5 mg/ml; Difco), or IFA. The emulsion
(total volume 0.15 ml) was injected s.c. at one site in the flank. Control
mice were
injected with PBS.
(xiii) Spinal cord injury. Mice were anesthetized, their spinal cords were
exposed by laminectomy at T12, and a force of 200 kdyn was placed for 1 s on
the
laminectomized cord using the Infinite Horizon spinal cord impactor (Precision

Systems and Instrumentation, Lexington, KY), a device shown to inflict a well-
calibrated contusive injury of the spinal cord.
(xiv) Assessment of functional recovery from spinal cord contusion.
Functional recovery from spinal cord contusion in mice was determined by
hindlimb locomotor performance. Recovery was scored by the Basso Mouse Scale
(BMS) open-field locomotor rating scale, a scale recently developed
specifically for
mice, with scores ranging from 0 (complete paralysis) to 9 (normal mobility)
(Engesser-Cesar et al., 2005). Blind scoring ensured that observers were not
aware
of the treatment received by each mouse. Twice a week locomotor activities of
mice
in an open field were monitored by placing the mouse for 4 min in the center
of a
circular enclosure (90 cm in diameter, 7 cm wall height) made of molded
plastic
with a smooth, non-slip floor. Before each evaluation the mice were examined
carefully for perineal infection, wounds in the hindlimbs, and tail and foot
autophagi a.
61

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
(xv) Stereotaxic injection of neural progenitor cells. Mice were
anesthetized and placed in a stereotactic device. The skull was exposed and
kept dry
and clean. The bregma was identified and marked. The designated point of
injection
was at a depth of 2 mm from the brain surface, 0.4 mm behind the bregma in the
anteroposterior axis, and 1.0 mm lateral to the midline. Neural progenitor
cells were
applied with a Hamilton syringe (5 x 105 cells in 3 I, at a rate of 1 1/min)
and the
skin over the wound was sutured.
(xvi) Neural progenitor cell culture. Cultures of adult neural progenitor
cells
(aNPCs) were obtained as previously described in Example 1.
(xvii) Co-culturing of neural progenitor cells and T cells. CD4+ T cells
were purified from lymph nodes of 8-week-old C57B16/J mice as previously
described (Kipnis et al., 2004). T cells were activated in RPMI medium
supplemented with L-glutamine (2 mM), 2-mercaptoethanol (5 x 10-5 M), sodium
pyruvate (1 mM), penicillin (100 IU/ml), streptomycin (100 g/ml),
nonessential
amino acids (1 m1/100 ml), and autologous serum 2% (v/v) in the presence of
mouse recombinant IL-2 (mrIL-2; 5 ng/ml) and soluble anti-CD28 and anti-CD3
antibodies (1 ng/ml). T cells were co-cultured (5 x 104 cells/well) with aNPCs
(5 x
104 cells/well) for 5 d on cover slips coated with Matrigel (BD Biosciences)
in 24-
well plates. The cultures were then fixed with 2.5% paraformaldehyde in PBS
for
30 min at room temperature and stained for neuronal markers.
(xviii) Immunohistochemistry. Mice subjected to SCI were re-anesthetized
14 or 60 days later and perfused with cold PBS. Their spinal cords were
removed,
postfixed with Bouin's fixative (75% saturated picric acid, 25% formaldehyde,
5%
glacial acetic acid; Sigma-Aldrich) for 48 h, and then transferred to 70%
Et0H. The
tissues were hydrated through a gradient of 70%, 95%, and 100% Et0H in xylene
and paraffin, and were then embedded in paraffin. For each stain, five tissue
sections, each 6 [im thick, were taken from each mouse. The paraffin was
removed
by successive rinsing of slides for 15 min with each of the following: xylene,
Et0H
100%, 95%, 70%, 50%, and PBS. Exposure of the slides to antigen was maximized
by heating them to boiling point in 10 mM sodium citrate pH 6.0 in a microwave
62

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
oven, then heating them at 20% microwave power for a further 10 min. The
slides
were blocked with 20% normal horse serum for 60 min prior to overnight
incubation at room temperature (GFAP, neurofilaments, and BDNF), or for 48 h
at
4 C (CD3), with the monoclonal antibody in 2% horse serum. We used rabbit anti-

mouse GFAP (1:200) (DakoCytomation, Glostrup, Denmark) for GFAP; rabbit
anti-neurofilament (1:200), low and high molecular weight (Serotec, Oxford,
UK)
for neurofilaments; and rat anti-human CD3 (1:50) (Serotec) for CD3. For BDNF
we used the monoclonal antibody chicken anti-human BDNF (1:100) (Promega,
Madison, WI) with 0.05% saponin.
After rinsing, sections were incubated for 1 h at room temperature with the
secondary antibody Cy3 donkey anti-rat (1:300) (Jackson ImmunoResearch
Laboratories, West Grove, PA) (staining for CD3), Cy3 donkey anti-chicken
(Jackson ImmunoResearch) (1:250) (staining for BDNF), or Cy3 donkey anti-
rabbit
(Jackson ImmunoResearch) (1:250) (staining for GFAP and neurofilaments). For
IB4 staining, sections were blocked for 1 h with 20% horse serum and then
incubated for 1 h at room temperature with Cy2-1B4 (1:50) (Sigma-Aldrich). All

sections were stained with Hoechst (1:2000) (Molecular Probes¨Invitrogen,
Carlsbad, CA). They were then prepared for examination under a Nicon E-600
fluorescence light microscope. Results were analyzed by counting the cells
(CD3-
labeled) in the site of injury, or by determination of the density (1B4-
labeled or
BDNF-labeled), or by measurement of the unstained area (GFAP, neurofilaments).

Each of the parameters was measured by an observer who was blinded to the
treatment received by the mice.
Example 2(1). Adult neural progenitor cells require local immune activity to
promote motor recovery
Our working hypothesis in this study was that the protective immune
response evoked at a site of injury by T-cell based immunization creates a
niche that
supports not only cell survival but also tissue repair. We further suspected
that a
local T-cell mediated immune response could attract exogenously delivered
aNPCs
63

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
and support their contribution to recovery. To test this hypothesis we
vaccinated
C57B1/6J mice, immediately after SCI, with the encephalitogenic peptide pMOG
35-55 (SEQ ID NO: 39) emulsified in CFA containing 0.5% Mycobacterium
tuberculosis (MOG/CFA), and 1 week later administered aNPCs via the
intracerebroventricular (i.c.v.) route. Mice subjected to this dual treatment
protocol
(MOG/CFA/aNPC) were compared to a control group of mice that were immunized
with the same MOG peptide emulsified in the same adjuvant but were not
transplanted with aNPCs and instead were injected i.c.v. with PBS
(MOG/CFA/PBS), or to mice that were injected with PBS and 'CFA (0.5%) and
transplanted i.c.v. with aNPCs (PBS/CFA/aNPC) or a control group of mice that
were injected with PBS/CFA and then injected i.c.v. with PBS (PBS/CFA/PBS). To

assess behavioral outcome after SCI we used the Basso motor score (BMS) rating

scale (Engesser-Cesar et al., 2005), in which 0 indicates complete paralysis
of the
hindlimbs and 9 denotes full mobility. The mean motor recovery (BMS) scores of
mice receiving the MOG/CFA/aNPC (4.21 1 0.45; all values are mean SEM)
were higher than those of mice treated with MOG/CFA/PBS. In mice treated with
PBS/CFA/aNPC, recovery was not better than in control mice treated with
PBS/CFA/PBS (1.5 0.27). A BMS of 4.21 indicates extensive movement of the
ankle and plantar placement of the paw (three animals showed, in addition,
occasional weight support and plantar steps), whereas a score of 1.5 indicates
ankle
movement ranging from slight to extensive. Mice treated with MOG/CFA/PBS
scored 2.71 0.5, a, significantly higher score than that of control
PBS/CFA/aNPC-
treated mice (1.5 th 0.4) or of control mice treated with PBS/CFA/PBS (Fig.
7A).
These results thus demonstrated synergistic interaction between the
administered
aNPCs and the T cell-based immune response. Failure of the transplanted aNPCs
to
improve motor recovery by themselves (i.e., in the absence of MOG/CFA
immunization) suggested that a site-specific immune response was necessary for

aNPC activity. Fig. 7B shows the BMS of individual mice in all examined groups

on day 28 postinjury. Because transplantation of aNPCs in the absence of
64

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
immunization did not improve recovery from SCI, this control group
(PBS/CFA/aNPC) was not included in subsequent experiments.
We have previously demonstrated that boosting of the amounts of T cells
needed for promoting recovery from SCI does not necessitate the use of
encephalitogenic peptides; weak agonists of encephalitogenic peptides are just
as
effective and do not carry the risk of inducing EAE. To test whether such
'safe'
vaccination could be utilized in combination with aNPCs transplantation we
used a
MOG-derived altered peptide ligand (pMOG 35-55 APL; 45D peptide, (SEQ ID
NO:40), in which aspartic acid is substituted for serine. Mice were vaccinated
with
the 45D peptide emulsified in CFA containing 2.5% Mycobacterium tuberculosis.
One week later the immunized mice were subjected to contusive SCI, and after
another week were transplanted i.c.v. with aNPCs. Increased motor activity (as

expressed by the BMS, mean SEM) was seen in these mice than in control mice
treated i.c.v with PBS/CFA/aNPC (4.11 0.27 compared to 1.94 0.22; Fig.
7C).
Without aNPC transplantation, immunization with peptide 45D in CFA resulted in
only a slight increase in motor recovery relative to the PBS-treated control
(2.57
0.24). Fig. 7D shows BMS values for individual mice on day 28 postinjury. The
above findings showed that the contribution of transplanted aNPCs to motor
recovery after contusive SCI could also be promoted by the use of a weak
agonist of
the encephalitogenic peptide.
To determine the phenotype and specificity of the T cells needed for
synergistic interaction with aNPCs we repeated the above experiments using
different immunization protocols. Incomplete Freund's adjuvant (IFA), unlike
CFA,
is free of bacteria and is known to elicit a Th2-like response to
encephalitogenic
peptides. We found that although immunization with the MOG analog (peptide
45D) emulsified in IFA had some beneficial effect, it showed no synergy with
subsequent transplantation of aNPCs (BMS of 3.2 0.76 for MOG/IFA/aNPC-
treated mice compared to 2.93 1.03 for MOG/IFA/PBS-treated mice and 2.07
0.53 in the PBS/CFA/PBS-treated mice; Fig. 7E). These findings suggest that
for

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
MOG/IFA/aNPC-treated mice, the preferred T cells for synergistic interaction
at the
injury site between vaccinated T cells and aNPCs are Thl.
To verify that specificity to CNS-antigens is required for a synergistic
effect
of vaccination and stern-cell transplantation, mice were immunized with the
nonself
protein ovalbumin (OVA) emulsified in CFA (containing 2.5% Mycobacterium
tuberculosis), and 1 week later were injected i.c.v. with aNPCs or with PBS as

control. Immunization with OVA/CFA resulted in a slight, nonsignificant
increase
in BMS, and implantation of aNPCs did not increase the BMS any further (2.43

1.78 for the OVA/CFA/aNPC-treated mice compared to 2.2 0.68 for
OVA/CFA/PBS-treated mice and 1.5 0.29 for PBS/CFA/PBS-treated control
group, Fig. 7F). Taking all of the above results together, the absence of a
beneficial
effect after aNPC transplantation suggests that synergy between T cells and
aNPCs
is a function of both the antigenic specificity and the phenotype of the T
cells.
Immune activation in the injured CNS, and specifically in the spinal cord,
has been a major focus of research attention in recent years (Schwartz and
Hauben,
2002). In some of the studies, T cell-based immune responses were shown to be
protective only if their intensity and duration were well regulated. Overly
strong
immunization yielded excessive immune activity, which neutralized the
potential
benefit of the immune response for the injured spinal cord and even had a
detrimental effect. We considered the possibility that if aNPCs home to the
site of
damage they can offset the negative effect of excessive immune activity and
thus
contribute to recovery. We therefore set out to determine whether
administration of
aNPCs can contribute to functional recovery even when the local immune
activity is
excessive. One week prior to SCI we immunized mice with MOG peptide 35-55
emulsified in CFA containing 2.5% Mycobacterium tuberculosis. Under conditions
in which motor recovery from SCI was worse after immunization with MOG/CFA
than after injection with PBS/CFA (BMS of 0.35 0.2 and 1.94 0.32,
respectively), we found that recovery was improved upon administration of
aNPCs
(BMS of 2.68 0.51; Figs. 7G, 7H). It thus appears that i.c.v. administration
of
66

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
aNPCs can contribute to functional recovery from SCI even when the local
adaptive
immune response is detrimental.
Example 2(2). Tissue integrity correlates with local immune activity
In an attempt to gain an insight into the mechanism underlying the apparent
synergy between aNPCs and resident immune cells, we examined whether any of
the injected aNPCs find their way to the injured spinal cord. We repeated the
experiment showed on Fig. 7A using GFP-labeled aNPCs. Staining with anti-GFP
antibodies revealed GFP+ cells in the parenchyma of the injured spinal cord
only in
mice that were treated with MOG-CFA/aNPC (Fig. 8). Injected GFP+ aNPCs could
be seen surrounding the epicenter of the lesion and laterally in the spinal
cord
parenchyma adjacent to the meninges as early as 7 days after SCI (Figs. 8A-
8C),
and could still be detected as late as 60 days after the injury, the last time
point
examined (Figs. 8D-8F).
One of the morphological features that characterize recovery from SCI is the
size of the lesion. To delineate the site of injury we stained longitudinal
sections of
the spinal cord with antibodies to glial fibrillary acidic protein (GFAP). We
assessed the lesion size by measuring the areas that were not stained by GFAP.
This
analysis disclosed that as early as 7 days after aNPCs were transplanted in
the
MOG/CFA-vaccinated rats, the averaged size of the site of injury was
significantly
smaller in mice that had received both vaccination and aNPC transplantation
than in
mice that had only been vaccinated or had received only aNPCs (Figs. 9A, 9B).
Next we examined whether the observed differences in the extent of recovery
could be correlated with local immunological changes. Sections of spinal cord
tissue were stained for markers of T cells (CD3) and accumulation of activated
microglia/macrophages (IB4) (Figs. 9C-9F). All sections were also stained with

Hoechst as a nuclear marker. Tissues were excised 7 days after cell
transplantation.
Staining with IB4 revealed fewer microglia/macrophages in mice that had
received
the dual treatment protocol (pMOG 35-55 in 0.5% CFA) than in the other
experimental groups (Figs. 9C, 9D). Quantitative analysis revealed
significantly
67

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
more CD3+ T cells in areas surrounding the site of injury in mice that had
received
the dual treatment than in MOG/CFA/PBS-treated or in PBS/CFA/PBS-treated
controls. Notably, immunization with the encephalitogenic pMOG 35-55 without
aNPC transplantation resulted in only slightly more CD3+ cells at the injured
site
than in the controls (Figs. 9E, 9F). It thus seems that transplantation of
aNPCs
modulated the local immune response at the injured site.
Both activated T cells and T cell-activated microglia can serve as sources of
growth factors such as brain-derived neurotrophic factor (BDNF). BDNF
immunoreactivity was more intense in the spinal cords of mice treated with
MOG/CFA/aNPC than in the other groups (Fig. 10A). Double staining for BDNF
and IB4 showed that the cellular source of BDNF in the injured site was the
microglial/macrophage population (Fig. 10B).
Recent studies have shown that noggin, a bone morphogenesis protein
(BMAD) inhibitor, can induce neuronal differentiation from aNPCs in the
injured
spinal cord (Setoguchi et al., 2004). This protein was also shown to be needed
to
provide a neurogenic environment in the subventricular zone. We therefore
assayed
noggin immunoreactivity in the various experimental groups, and found that it
was
significantly increased in mice that received the dual treatment protocol, but
was
unaffected by MOG immunization alone and was slightly decreased by aNPC
transplantation alone (Fig. 10C). As in the case of BDNF produced in the
injured
site, noggin was also localized to IB4+ cells (Fig. 10D).
Example 2(3). Local differentiation of endogenous stem cells
The above results raised an important question: can a T cell-based
vaccination, when given in combination with aNPC transplantation, create
conditions favorable for neuronal differentiation of endogenous or exogenous
aNPCs? To examine this possibility, we repeated the experiment described in
Fig.
7, while also injecting the cell-proliferation marker BrdU twice daily for 3
days,
starting on day 7 after aNPC transplantation (i.e., 14 days after SCI).
Staining for
BrdU and the early differentiation marker doublecortin (DCX) 7 days after the
last
68

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
BrdU injection disclosed significantly more newly formed neurons in the mice
that
had received the dual treatment (Figs. 11A-11E). Fig. 11B demonstrates the
distribution of DCX+ cells in the environment/vicinity of the injured site.
Staining
for the combination of BrdU and GFP and of GFP and DCX revealed virtually no
double-positive cells, indicating that most of the DCX+ cells in the injured
spinal
cord had originated from endogenous aNPCs. Notably, vaccination without aNPC
transplantation did not increase the formation of new neurons in the injured
spinal
cord.
Example 2(4). Interaction of adult neural progenitor cells and T cells in
vitro
The above results indicated that cross-talk between immune cells and aNPCs
was taking place at the site of injury. We showed hereinabove that microglia
pre-
activated with the Thl- and Th2-associated cytokines, IFN-y and IL-4,
respectively,
can induce neuronal differentiation from aNPCs. We therefore sought to
determine
whether direct interaction between aNPCs and T cells would also result in an
altered
pattern of aNPC differentiation. To address this question, we activated CD4+ T

cells in vitro by a cognate protocol (with anti-CD3 antibodies, anti-CD28
antibodies, and IL-2) for 24 h and then allowed their activation to continue
in co-
cultures with aNPCs in a transwell culture system. As controls we used
cultures of
aNPCs alone (in the presence of anti-CD3 antibodies, anti-CD28 antibodies and
IL-
2) or aNPCs cultured with CD4+ T cells in a resting state (supplemented with
IL-2
only). After 5 days in culture the aNPCs in the lower chamber were fixed and
analyzed for the appearance of newly formed neurons. Staining for the early
neuronal marker P-III-tubulin revealed a dramatic effect of T cells on
neuronal
differentiation (Fig. 12A). Compared to control cultures of aNPCs alone, in
which
only about 7% of the cells were positive for P-III-tubulin, approximately 90%
of the
cells expressed p-III-tubulin in co-cultures of aNPCs with activated T cells
(Figs.
12A, 12B). Notably, p-III-tubulin staining showed a 3-fold increase (18%)
relative
to the control in co-cultures of aNPCs with nonactivated T cells. It thus
seems that
T cells induce neuronal differentiation via a soluble factor. Fig. 12B shows
69

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
representative images of (3-111 tubulin-stained cultures of aNPCs alone
(control) or
of co-cultures with activated T cells. These findings imply that T cell-
derived
soluble factors might trigger one of the pathways of neuronal differentiation.
To exclude the possibility that the T cells had affected NPC differentiation
as
a consequence of encountering NPC-derived compounds, and to further
substantiate
our finding that the observed effect was caused by T cell-derived soluble
substances, we allowed aNPCs to differentiate in the presence of medium
conditioned by activated T cells. After 5 days, staining of these cultures for
13-III-
tubulin revealed similar results to those obtained in the co-culture system
(Fig
12C), confirming that neuronal differentiation was induced by resting T cells
and
even more by the activated T cells. In addition to their effect on the numbers
of
differentiating cells, the T cell-derived soluble factors also evidently
affected the
cellular morphology, as manifested in the branched, elongating f3-III tubulin-
labeled
fibers (Fig. 12D).
We next sought to determine whether the T cell-induced neurogenesis was
mediated by cytokines secreted by activated T cells. aNPCs were cultured in
the
presence of different concentrations of the characteristic T-cell derived
cytokines
IFN-y and IL-4. Analysis revealed that IFN-y, at concentrations as low as 1
ng/ml,
could induce an increase in 13-III-tubulin expression after 5 days in culture
(Fig
12E). In experiments described herein, we found that brief exposure to IFN-y
(24 h)
was not sufficient to attain such an effect. It should be noted, however, that
the
morphology of the 13-III-tubulin-expressing cells in IFN-y-supplemented
cultures
was less developed than that seen in aNPCs cultured with activated T cells or
in T
cell-conditioned medium. In contrast to the effect of IFNI, no change in 13-
III-
tubulin expression was observed in aNPCs treated with IL-4.
These findings suggested that IFN-y, unlike IL-4, could account in part for
the T cell-induced neurogenesis. Even the effect of IFN-y, however, was
limited
relative to that of the T cells or to the T cell-derived soluble factors. PCR
analysis of

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
expression of the IFN-y receptor-1 on aNPCs disclosed that this receptor is
expressed by aNPCs under all of the conditions examined here (data not shown).

Activation of the Notch pathway is essential for maintenance of aNPCs, and
blockage of this pathway and its downstream transcription factors of the Hes
gene
family underlie the first events in neuronal differentiation. To determine
whether T
cell-mediated neuronal differentiation induces changes in Notch signaling, we
looked for possible changes in expression of Hes genes in aNPCs following
their
interaction with T cell-derived substances. Real-time PCR disclosed that
relative to
control cultures, aNPCs cultured for 24 h in the presence of T cell-
conditioned
medium underwent a five-fold decrease in Hes-5 expression (Fig. 12F). Thus,
differentiation induced by T cells appears to involve inhibition of the Notch
pathway. Expression of Notch 1-4 by aNPCs was not altered in the presence of T

cell-conditioned medium, indicating that the inhibition could not be
attributed to
changes in Notch expression (data not shown).
Our observation that aNPCs express an IFN-y receptor, taken together with
recent studies showing that these cells express immune-related molecules such
as B-
7 (Imitola et al., 2004b) and CD44 (Pluchino et al., 2003), known to
participate in
the dialog between T cells and antigen-presenting cells (APCs), prompted us to

examine whether aNPCs could affect T-cell function. First we examined the
effects
of aNPCs on proliferation of CD4+ T cells by assaying [3H]thymidine
incorporation
by the T cells. Co-culturing of T cells with aNPCs and APCs (lethally
irradiated
splenocytes) for 3 days resulted in a significant dose-dependent inhibition of
T-cell
proliferation (Fig. 13A). It is important to note that under these conditions
there was
only limited proliferation of aNPCs. To determine whether the inhibitory
effect on
T-cell proliferation is mediated by a soluble factor or requires cell¨cell
contact, we
utilized the transwell system, plating aNPCs in the upper well. Co-culturing
of T
cells and aNPCs in the same well resulted in a two-fold reduction in T-cell
proliferation, but this effect was diminished when the two cell populations
were
separated in the transwell. It thus seems that cell¨cell contact is necessary
for
aNPCs to inhibit T-cell proliferation. To determine whether aNPCs could affect
the
71

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
production of cytokines by T cells, we measured the concentrations of six
inflammatory cytokines in the media of co-cultured aNPCs and T cells. The
concentrations of IL-12, IFN-y, and TNF-a were similar in T-cell cultures with
and
without aNPCs, but the concentration of IL-10 was slightly increased (by 40%)
in
the co-culture. Relative to T-cell cultures alone, the concentration of IL-6
was
twofold higher in the co-culture with aNPCs, and a remarkable difference was
seen
in MCP-1 concentration, which was higher by two orders of magnitude in the co-
culture (Fig. 13C). Taken together, these results indicate that aNPCs can act
directly
on T cells, inhibiting their proliferative activity and changing their
cytokine/
chemokine production profile.
Discussion
Local interaction between immune cells and aNPCs underlies functional
recovery. In the present study we combined two different therapeutic
approaches for
SC': T cell-based vaccination and transplantation of neural progenitor cells
into the
CSF. Each of these approaches has been shown to be potentially capable of
promoting functional recovery from SCI; we show here that when combined, they
operate in synergy. Our experiments, both in vivo and in vitro, demonstrated
tthat
cross talk between immune cells and aNPCs can take place at the lesion site.
The
vaccination elicits a local immune response, which, if well controlled,
provides the
cellular and molecular elements needed to attenuate degeneration and promote
repair. The same response also plays a role in recruiting aNPCs to the injured
site
and creating niche-like compartments that support neurogenesis from endogenous

aNPCs. The interaction between aNPCs and immune cells was found to be
reciprocal: aNPCs could modulate the postinjury immune activity, ensuring
functional recovery even under conditions of excessive immune activity (which,
in
the absence of aNPCs, have a detrimental effect on recovery).
72

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
EXAMPLE 3
T-cell Based Vaccination Restores Cognition, Removes Plaques, and
Induces Neurogenesis in a Mouse Model of Alzheimer's Disease
Accumulation of P-amyloid deposition (AP), neuronal loss, cognitive
decline, and microglial activation, are characteristic features of Alzheimer's
disease
(AD). Using AD double-transgenic mice expressing mutant human genes encoding
presenilin 1 and chimeric mouse/human amyloid precursor protein, we show that
vaccination with glatiramer acetate prevented and restored cognitive decline,
assessed by performance in a Morris water maze (MWM). The vaccination
modulated microglial activation, eliminated plaque formation, and induced
neuronal
survival and neurogenesis. In vitro, AP-activated microglia impeded
neurogenesis
from adult neural stem/progenitor cells. This was counteracted by IL-4, and
more so
when IFN-y was added, but not by IFN-y alone.
Materials and Methods
(xLv) Animals. Nineteen adult double-transgenic APPK670N, M671L + PS 4E9
mice of the B6C3-Tg (APPswe, PSEN1dE9) 85Dbo/J strain were purchased from
The Jackson Laboratory (Bar Harbor, ME) and were bred and maintained in the
Animal Breeding Center of The Weizmann Institute of Science. A11 animals were
handled according to the regulations formulated by the Weizmann Institute's
Animal Care and Use Committee. Tg AD mice were produced by co-injection of
chimeric mouse/human APPswe (APP695 [humanized AP domain] harboring the
Swedish [K594M/N595L] mutation) and human PS1dE9 (deletion of exon 9)
vectors controlled by independent mouse prion protein promoter (MoPrP)
elements,
as described (Borchelt et al., 1997).
(xx) Reagents. Recombinant mouse IFN-y and IL-4 (both containing
endotoxin at a concentration below 0.1 ng/lig cytokine) were obtained from R&D

Systems (Minneapolis, MN). P-amyloid peptides [amyloid protein fragment 1-40
73

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
and 1-42 (A31-4011-42)] were purchased from Sigma-Aldrich, St. Louis, MO. The
AP peptides were dissolved in endotoxin-free water, and Af3 aggregates were
formed by incubation of A(3, as described (Ishii et al., 2000).
(xxi) Genotyping. All mice used in this experiment were genotyped for the
presence of the transgenes by PCR amplification of genomic DNA extracted from
1-cm tail clippings (Jankowsky et al., 2004). Reactions contained four
primers: one
anti-sense primer-matching sequence within the vector that is also present in
mouse
genomic PrP (5'-GTG GAT ACC CCC TCC CCC AGC CTA GAC C) (SEQ ID
NO:41); a second sense primer specific for the genomic PrP coding region
(which
was removed from the MoPrP vector) (5'-CCT CTT TGT GAC TAT GTG GAC
TGA TGT CGG) (SEQ ID NO:42); and two sense and anti-sense primers specific
for the PS1 transgene cDNA (PS1-a: 5'-AAT AGA GAA CGG CAG GAG CA
(SEQ ID NO:43), and PS1-b: 5'-GCC ATG AGG GCA CTA ATC AT) (SEQ ID
NO:44). All reactions give a 750-bp product of the endogenous PrP gene as a
control for DNA integrity and successful amplification; PS1 transgene-positive
samples have an additional band at approximately 608 bp.
(xxii) Glatiramer acetate vaccination. Each mouse was subcutaneously
injected five times with a total of 100 [tg of glatiramer acetate (GA (TV-
5010), MW
13.5-18.5 kDa, average 16 kDa, Teva Pharmaceutical Industries Ltd., Petach
Tikva,
Israel), emulsified in 200 IA PBSx 1, from experimental day 0 until day 24,
twice
during the first week and once a week thereafter.
(xxiii) Behavioral testing. Spatial learning/memory was assessed by
performance on a hippocampus-dependent visuo-spatial learning task in the
Morris
water maze (MWM) (Morris, 1984). Mice were given four trials per day on 4
consecutive days, during which they were required to find a hidden platform
located
1.5 cm below the water surface in a pool 1.4 m in diameter. Within the testing

room, only distal visuo-spatial cues for location of the submerged platform
were
available. The escape latency, i.e., the time required by the mouse to find
the
platform and climb onto it, was recorded for up to 60 s. Each mouse was
allowed to
remain on the platform for 30 s and was then moved from the maze to its home
74

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
cage. If the mouse did not find the platform within 60 s, it was placed
manually on
the platform and returned to its home cage after 30 s. The interval between
trials
was 300 s. On day 5 the platform was removed from the pool and each mouse was
tested by a probe trial for 60 s. On days 6 and 7 the platform was placed at
the
quadrant opposite the location chosen on days 1-4, and the mice were then
retrained
in four sessions per day. Data were recorded using an EthoVision automated
tracking system (Noldus).
(xxiv) Administration of BrdU and tissue preparation. BrdU was dissolved
by sonication in PBS and injected i.p. into each mouse (50 mg/kg body weight;
1.25
mg BrdU in 200 1.11 PBS xl). Starting from experimental day 22 after the first
GA
vaccination, BrdU was injected i.p. twice daily, every 12 h for 2.5 days, to
label
proliferating cells. Three weeks after the first BrdU injection the mice were
deeply
anesthetized and perfused transcardially, first with PBS and then with 4%
paraformaldehyde. The whole brain was removed, postfixed overnight, and then
equilibrated in phosphate-buffered 30% sucrose. Free-floating 30- m sections
were
collected on a freezing microtome (Leica SM2000R) and stored at 4 C prior to
immunohistochemistry.
(xxv) Neural progenitor cell culture. Coronal sections (2 mm thick) of tissue
containing the subventricular zone of the lateral ventricle were obtained from
the
brains of adult C57B1/6J mice. The tissue was minced and then incubated for
digestion at 37 C, 5% CO2 for 45 min in Earle's balanced salt solution
containing
0.94 mg/ml papain (Worthington, Lakewood, NJ) and 0.18 mg/ml of L-cysteine and

EDTA. After centrifugation at 110 x g for 15 min at room temperature, the
tissue
was mechanically dissociated by pipette trituration. Cells obtained from
single-cell
suspensions were plated (3500 cells/cm2) in 75-cm2 Falcon tissue-culture
flasks
(BD Biosciences, San Diego, CA), in NPC-culturing medium [Dulbecco's modified
Eagles's medium (DMEM)/F12 medium (Gibco/Invitrogen, Carlsbad, CA)
containing 2 mM L-glutamine, 0.6% glucose, 9.6 1..tg/m1 putrescine, 6.3 ng/ml
progesterone, 5.2 ng/ml sodium selenite, 0.02 mg/ml insulin, 0.1 mg/ml
transferrin,
2 p.g/m1 heparin (all from Sigma-Aldrich, Rehovot, Israel), fibroblast growth
factor-

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
2 (human recombinant, 20 ng/ml), and epidermal growth factor (human
recombinant, 20 ng/ml; both from Peprotech, Rocky Hill, NJ)]. Spheres were
passaged every 4-6 days and replated as single cells. Green fluorescent
protein
(GFP)-expressing NPCs were obtained as previously described (Pluchino et al.,
2003).
(xxvi) Primary microglial culture. Brains from neonatal (P0¨P1) C57B1/6J
mice were stripped of their meninges and minced with scissors under a
dissecting
microscope (Zeiss, Stemi DV4, Germany) in Leibovitz-15 medium (Biological
Industries, Kibbutz Beit Ha-Emek, Israel). After trypsinization (0.5% trypsin,
10
min, 37 C/5% CO,), the tissue was triturated. The cell suspension was washed
in
culture medium for glial cells [DMEM supplemented with 10% fetal calf serum
(FCS; Sigma-Aldrich, Rehovot), L-glutamine (1 mM), sodium pyruvate (1 mM),
penicillin (100 U/ml), and streptomycin (100 mg/ml)] and cultured at 37 C/5%
CO2
in 75-cm2 Falcon tissue-culture flasks (BD Biosciences) coated with poly-D-
lysine
(PDL) (10 mg/ml; Sigma-Aldrich, Rehovot) in borate buffer (2.37 g borax and
1.55
g boric acid dissolved in 500 ml sterile water, pH 8.4) for 1 h, then rinsed
thoroughly with sterile, glass-distilled water. Half of the medium was changed
after
6 h in culture and every 2nd day thereafter, starting on day 2, for a total
culture time
of 10-14 days. Microglia were shaken off the primary mixed brain glial cell
cultures (150 rpm, 37 C, 6 h) with maximum yields between days 10 and 14,
seeded (105 cells/m1) onto PDL-pretreated 24-well plates (1 ml/well; Corning),
and
grown in culture medium for microglia [RPMI-1640 medium (Sigma-Aldrich)
supplemented with 10% FCS, L-glutamine (1 mM), sodium pyruvate (1 mM), 13-
mercaptoethanol (50 mM), penicillin (100 U/ml), and streptomycin (100 mg/ml)].
The cells were allowed to adhere to the surface of a PDL-coated culture flask
(30
min, 37 C/5% CO2), and non-adherent cells were rinsed off.
(xxvii) Co-culturing of mouse neural progenitor cells and mouse
microglia. Cultures of treated or untreated microglia were washed twice with
fresh
NPC-differentiation medium (same as the culture medium for NPCs but without
growth factors and with 2.5% FCS) to remove all traces of the tested reagents,
then
76

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
incubated on ice for 15 min, and shaken at 350 rpm for 20 min at room
temperature.
Microglia were removed from the flasks and immediately co-cultured (5 x 104
cells/well) with NPCs (5 x 104 cells/well) for 10 days on cover slips coated
with
MatrigelTM (BD Biosciences) in 24-well plates, in the presence of NPC
differentiation medium. The cultures were then fixed with 2.5%
paraformaldehyde
in PBS for 30 min at room temperature and stained for neuronal and glial
markers.
(xxviii) Immunocytochemistry and inununohistochendsny. Cover slips
from co-cultures of NPCs and mouse microglia were washed with PBS, fixed as
described above, treated with a permeabilization/blocking solution containing
10%
FCS, 2% bovine serum albumin, 1% glycine, and 0.1% Triton X-100 (Sigma-
Aldrich, Rehovot), and stained with a combination of the mouse anti-tubulin 13-
III-
isoform C-terminus antibodies (13-III-tubulin; 1:500; Chemicon, Temecula, CA)
and
CD1lb (MAC1; 1:50; BD¨Pharmingen, Franklin Lakes, NJ).
For BrdU staining, sections were washed with PBS and incubated in 2N HC1
at 37 C for 30 min. Sections were blocked for 1 h with blocking solution (PBS
containing 20% normal horse serum and 0.1% Triton X-100, or PBS containing
mouse immunoglobulin blocking reagent obtained from Vector Laboratories
(Burlingame, CA)).
For immunohistochemistry, tissue sections were treated with a
permeabilization/blocking solution containing 10% FCS, 2% bovine serum
albumin, 1% glycine, and 0.05% Triton X-100 (Sigma-Aldrich, St. Louis). Tissue

sections were stained overnight at 4 C with specified combinations of the
following
primary antibodies: rat anti-BrdU (1:200; Oxford Biotechnology, Kidlington,
Oxfordshire, UK), goat anti-DCX [doublecortin] (1:400; Santa Cruz
Biotechnology), and mouse anti-NeuN [neuronal nuclear protein] (1:200;
Chemicon, Temecula, CA). Secondary antibodies were FITC-conjugated donkey
anti-goat, Cy-3-conjugated donkey anti-mouse, and Cy-3- or Cy-5-conjugated
donkey anti-rat (1:200; Jackson ImmunoResearch, West Grove, PA). For labeling
of microglia we used either CD11 b (MAC1; 1:50; BD¨Pharmingen) or FITC-
conjugated Bandeiraea simplicifolia isolectin B4 (IB4, 1:50; Sigma-Aldrich,
77

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Rehovot). To detect expression of cell-surface
proteins we used anti-MHC-
II Abs (rat, clone IBL-5/22; 1:50; Chemicon, Temecula, CA). To detect
expression
of human A13 we used anti-A13 (human amino-acid residues 1-17) (mouse, clone
6E10, Chemicon, Temecula, CA). Expression of IGF-I was detected by goat anti-
IGF-I Abs (1:20; R&D Systems). Expression of TNF-oc was detected by goat anti-
TNF-st Abs (1:100; R&D Systems). T cells were detected with anti-CD3
polyclonal
Abs (rabbit, 1:100; DakoCytomation, CA). Propidium iodide (1 1.1g/m1;
Molecular
Probes, Invitrogen, Carlsbad, CA) was used for nuclear staining.
Control sections (not treated with primary antibody) were used to distinguish
specific staining from staining of nonspecific antibodies or autofluorescent
components. Sections were then washed with PBS and coverslipped in polyvinyl
alcohol with diazabicylo-octane as anti-fading agent.
(xxix) Quantification and stereological counting procedure. For
microscopic analysis we used a Zeiss LSM 510 confocal laser-scanning
microscope
(40x magnification). For experiments in vitro we scanned fields of 0.053 mm2
(n =
8-16 from at least two different coverslips) for each experimental group. For
each
marker, 500-1000 cells were sampled. Cells co-expressing GFP and 13-III-
tubulin
were counted.
For in-vivo experiments, the number of A13+ plaques and CD11b+/IB-4+
microglia in the hippocampus were counted at 300- m intervals from 6-8 coronal
sections (30 !um) from each mouse. Neurogenesis in the DG was evaluated by
counting of premature neurons (DCX+), proliferating cells (BrdU+), and newly
formed mature neurons (BrdU+/NeuN+) in six coronal sections (30 vun) from each

mouse. Specificity of BrdLT+/NeuN+ co-expression was assayed using the
confocal
microscope (LSM 510) in optical sections at 1-pm intervals/. Cell counts,
numbers
of A13+ plaques, plaque areas, and intensity of NeuN staining per unit area in
the DG
were evaluated automatically using Image-Pro Plus 4.5 software (Media
Cybernetics).
78

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
(xxx) Statistical analysis. MWM behavior scores were analyzed using 3-way
ANOVA, with treatment group and trial block as sources of variation, was used
to
evaluate the significance of differences between mean scores during
acquisition trial
blocksin the MWM. When the P-value obtained was significant, a pairwise
Fisher's
least-significant-difference multiple comparison test was run to determine
which
groups were significantly different.
The in-vitro results were analyzed by the Tukey¨Kramer multiple
comparisons test (ANOVA) and are expressed as means SEM. In-vivo results
were analyzed by Student's t-test or 1-way ANOVA and are expressed as means
SEM.
Example 3(1). T cell-based vaccination counteracts cognitive decline in AD
We examined the effect of GA in AD double-transgenic mice (Tg mice)
expressing a mutant human presenilin 1 gene (PS1dE9) and a chimeric
mouse/human amyloid precursor protein (APPswe), leading to learning/memory
impairment and accumulation of Ai3 plaques mainly in the cortex and the
hippocampus, both characteristic features of early-onset familial AD (Borchelt
et
al., 1997). Expression of both transgenes in each mouse was verified by PCR
amplification of genomic DNA. APP/PS1 Tg mice aged approximately 8 months
were then vaccinated subcutaneously with GA (n = 6) twice during the first
week
and once a week thereafter. Age-matched Tg mice (n = 7) and non-Tg littermates

that did not carry the transgenes (n= 6), were not treated and served as
untreated Tg
and wild-type non-Tg controls, respectively. Five weeks after the first GA
injection
all mice were assessed in a Morris water maze (MWM) for cognitive activity, as
reflected by performance of a hippocampus-dependent spatial learning/memory
task
(reviewed in van Praag et al., 2000). The MWM performance of the untreated Tg
mice was significantly worse, on average, than that of the age-matched non-Tg
littermates (Fig. 14). However, the performance of Tg mice that were
vaccinated
with GA was= superior to that of the untreated Tg mice and did not differ
significantly from that of their non-Tg littermates (Fig. 14), suggesting that
the GA
79

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
vaccination had prevented further cognitive loss and even reversed part of the

earlier functional deficit. Cognitive losses or improvements were manifested
in both
the acquisition and the reversal tasks (Figs. 14A-14C).
Example 3(2). T cell-based vaccination modulates the immune activity of
microglia, eliminates 13-amy1oid plaque formation, supports neuronal survival
and induces neurogenesis
The above results prompted us to examine the possibility that the observed
arrest and reversal of cognitive loss was related to reduction of AP plaques
and
survival of neurons in the hippocampus. Staining of brain cryosections from Tg
mice with antibodies specific to human AP disclosed numerous plaques in the
untreated Tg mice but very few in those vaccinated with GA (Fig. 15A). No
plaques
were seen in their respective non-Tg littermates (Fig. 15A). Examination of
NeuN
immunoreactivity disclosed loss of neurons in the untreated Tg mice but
preservation of neurons in the GA-vaccinated Tg mice (Fig. 15A).
Activated microglia are known to play a role in the pathogenesis of AD. We
have shown in the Examples hereinbefore that, unlike microglia seen in
association
with inflammatory and neurodegenerative diseases, the microglia associated
with
neural tissue survival express MTIC-II, produce IGF-1, and express little or
no TNF-
a. We therefore examined brain sections from GA-vaccinated and untreated Tg
mice for the presence of microglia that stain positively for CD1 lb or TNF-a
(markers of activation associated with a cytotoxic inflammatory phenotype).
The
presence of plaques was found to be correlated with the appearance of CD1 lb+
microglia (Fig. 15B) expressing TNF-a (Fig. 15C and Movie S1 (prepared by the
inventors but not shown here) that depicts a 3-D reconstruction of an AP
plaque and
CD11b+ microglia expressing TNF-a. This movie presents a representative 3-D
confocal image of a microglial cell embedded within an Ap plaque in the
hippocampus of an untreated Tg mouse shown in Fig. 15C, in which the high TNF-
a-immunoreactivity and engulfed AP in the cytoplasm can be noted), and was

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
abundant in the untreated Tg mice. Significantly fewer CD1111+ microglia were
detectable in the GA-vaccinated Tg mice (Fig. 15B). Staining with anti-MHC-II
antibodies disclosed that in the untreated Tg mice almost no microglia
expressed
MHC-H (indicating their inability to act as APCs; data not shown), whereas in
the
GA-vaccinated Tg mice most of the microglia adjacent to residual AP+ plaques
expressed MHC-II, and hardly any of them expressed TNF-a (Fig. 15D and Movie
S2 (prepared by the inventors but not shown here) that depicts 3-D
reconstruction of
an AP¨immunoreactivity associated with MHC-II+ microglia in a GA-vaccinated
Tg mouse. This movie also shows a representative 3-D confocal image of
microglia
shown in Fig. 15D, expressing marginal levels of TNF-a and high levels of Ml-
IC-
11). These latter microglia also expressed IGF-I (Fig. 15E and Movie S3
(prepared
by the inventors but not shown here) that depicts a 3-D reconstruction of a
microglial cell co-expressing IGF-1 and MHC-111. This movie shows a
representative 3-D confocal image of MIC-II+ microglia from the IGF-I-
expressing
GA-vaccinated Tg mouse shown in Fig. 15E.), indicating their potential for
promoting neuroprotection and neurogenesis and for beneficially affecting
learning
and memory. All of the MHC-II+ cells were co-labeled with IB4, identifying
them
as microglia (data not shown). It is important to note that the CD11b+
microglia
(seen mainly in the untreated Tg mice) showed relatively few ramified
processes,
whereas such processes were abundant in the MHC-II+ microglia in the GA-
vaccinated Tg mice, giving them a bushy appearance (depicted in Movies S1 and
S2, not shown).
In addition, unlike in the untreated Tg mice, in the GA-vaccinated Tg mice
numerous T cells (identified by anti-CD3 antibodies) were seen in close
proximity
to MHC-II+ microglia. Any AP-immunoreactivity seen in these mice appeared to
be
in association with MHC-II+ microglia, creating an immune synapse with CD3+ T
cells (Fig. 15F and Movie S4 (prepared by the inventors but not shown here)
that
depicts a 3-D reconstruction of an Ap plaque associated with CD3+ cells (T
cells) in
close proximity to MHC-II+ microglia. This movie depicts a representative 3-D
confocal image of MHC-II+ microglia from the GA-vaccinated Tg mouse shown in
81

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Fig. 15F, with an immunological synapse between a CD3+ cell and a complex of
MHC-II and Ap.).
Quantitative analysis confirmed that mice vaccinated with GA showed
significantly fewer plaques than untreated Tg mice when examined 6 weeks later
(Fig. 15G), and that the area occupied by the plaques was significantly
smaller than
in their age-matched untreated counterparts (Fig. 15H). In addition, GA-
vaccinated
Tg mice showed significantly fewer CD1113+ microglia and significantly more
intense staining for NeuN than their corresponding groups of untreated Tg mice

(Figs. 151, 15J).
Because MHC-II-expressing microglia are also associated with neurogenesis
in vitro, we examined the same sections for the formation of new neurons in
the
dentate gyrus (DG) of the hippocampus. This was possible because all mice had
been injected with BrdU, a marker of proliferating cells, 3 weeks before
tissue
excision. Quantitative analysis disclosed significantly more BrdU+ cells in GA-

vaccinated Tg mice (Fig. 16A) than in their untreated counterparts. In
addition,
compared to the numbers of newly formed mature neurons (BrdU+/NeuN+) in their
respective non-Tg littermates the numbers were significantly lower in the
untreated
Tg group, but were similar in the vaccinated group, indicating that the
neurons had
been at least partially restored by the GA vaccination (Fig. 16B). Analysis of
corresponding sections for doublecortin (DCX), a useful marker for analyzing
the
absolute number of newly generated pre-mature neurons in the adult DG,
disclosed
that relative to the non-Tg littennates there were significantly fewer DCX+
cells in
the DGs of untreated Tg mice, and slightly but significantly more in the DGs
of Tg
mice vaccinated with GA (Fig. 16C). Confocal micrographs illustrate the
differences in the numbers of BrdU+/NeuN+ cells or of DCX+ cells and their
dendritic processes between non-Tg littermates, untreated Tg mice, and GA-
vaccinated Tg mice (Fig. 16D). The results showed that neurogenesis was indeed

more abundant in the GA-vaccinated mice than in untreated Tg mice.
Interestingly,
however, in both untreated and GA-vaccinated Tg mice the processes of the DCX+-

stained neurons in the subgranular zone of the DG were short, except in those
GA-
82

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
vaccinated mice in which the DCX+ cells were located adjacent to MHC-II+
microglia (Fig. 16E).
Example 3(3). Aggregated f3-amyloid induces microglia to express a phenotype
that blocks neurogenesis, and the blocking is counteracted by IL-4
The in-vivo results presented above point to a relationship between arrested
neurogenesis, aggregated A.13, and the phenotype of the activated microglia.
To
determine whether aggregated Af3-activated microglia block neurogenesis, and
whether T cell-derived cytokines can counteract the inhibitory effect, we co-
cultured GFP-expressing NPCs with microglia that had been pre-incubated for 48
h
in their optimal growth medium in the presence or absence of aggregated Al3
peptide 1-40/1-42 (A0(1-40/1-42); 5 iuM) and subsequently treated with IFN-y
(10
ng/ml), or with 1L-4 (10 ng/ml) together with IFN-y (10 ng/ml), for an
additional 48
h. Growth media and cytokine residues were then washed off, and each of the
treated microglial preparations was freshly co-cultured with dissociated NPC
spheres on coverslips coated with MatrigelTM in the presence of
differentiation
medium (Fig. 17A). Expression of GFP by NPCs confirmed that any
differentiating
neurons seen in the cultures were derived from the NPCs rather than from
contamination of the primary microglial culture. After 10 days we could
discern
GFP-positive NPCs expressing the neuronal marker P-III-tubulin ([3IIIT) (Figs.
17B, 17C). No r3IIIT+ cells were seen in microglia cultured without NPCs.
Significantly fewer GFP+/PIIIT+ cells were seen in control NPCs cultured
without
microglia (control). In co-cultures of NPCs with microglia previously
activated by
incubation with IFN-y (10 ng/ml), however, the increase in numbers of
GFP+43IIIT+
cells was dramatic. In contrast, microglia activated by aggregated A13(1-40)
(5 PM)
blocked neurogenesis and decreased the number of NPCs. This negative effect
was
not exhibited by microglia activated by 5 M A13(l_42) (data not shown).
Interestingly, the addition of 1L-4 (10 ng/ml) to microglia pre-treated with
aggregated Ar3(l_40) partially counteracted the adverse effect of the
aggregated Al3
83

CA 02588908 2007-05-29
WO 2006/057003
PCT/1L2005/001275
on NPC survival and differentiation, with the result that these microglia were
able
to induce NPCs to differentiate into neurons (Fig. 17D). It thus appears that
aggregated A,11(l_40) impaired the ability to support neurogenesis, and that
its effect
could be counteracted to some extent by IL-4 and more strongly by the
combination
of IL-4 and IFN-y.
Discussion
In this study of APP/PS1 double-transgenic AD mice suffering from decline
in cognition and accumulation of AP plaques, a T cell-based vaccination, by
altering the microglial phenotype, ameliorated cognitive performance, reduced
plaque formation, rescued cortical and hippocampal neurons, and induced
hippocampal neurogenesis.
EXAMPLE 4
Combination of glatiramer acetate vaccination and stem cells in an
animal model of amyotrophic lateral sclerosis (ALS)
Materials and methods
(xxxi) Animals. Transgenic mice overexpressing the defective human
mutant SOD1 allele containing the G1y934A1a (G93A) gene (B6SJL¨TgN
(SOD1¨G93A)1Gur (herein "ALS mice") were purchased from The Jackson
Laboratory (Bar Harbor, ME, USA).
(xxxii) Immunization. Adult mice were immunized with Cop-1, 100 j.ig in
200111 PBS s.c.
(xxxiii) Neural progenitor cell culture. Cultures of adult neural progenitor
cells (aNPCs) were obtained as described in orevious examples
(xxxiv) Stereotaxic injection of neural progenitor cells. Mice were
anesthetized a week after the first immunization and placed in a stereotactic
device.
The skull was exposed and kept dry and clean. The bregma was identified and
marked. The designated point of injection was at a depth of 2 mm from the
brain
84

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
surface, 0.4 mm behind the bregma in the anteroposterior axis, and 1.0 mm
lateral
to the midline. Neural progenitor cells were applied with a Hamilton syringe
(5 x
105 cells in 3 p.1, at a rate of 1 ill/min) and the skin over the wound was
sutured.
(xxxv) Motor dysfunction. Motor dysfunction of the mice was evaluated
two weeks twice a week Cop-1, thereafter they received a weekly injection of
Cop-
1. The stein cells were given into the CSF: 500, 000 cells (single injection
of adult
neural stem cells).
The experiment was carried out in order to explore whether administration of

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Mice from each of the groups were weighted (twice a week) and examined
routinely for vital signs, and for signs of motor dysfunction. The results
obtained in
Fig. 44 show the probability of survival of each group of ALS mice. The
results
show that the combined treatment of Cop-1 vaccination and NPC results in
increased survival of ALS mice.
EXAMPLE 5
Neurogenesis and neuroprotection induced by peripheral
immunomodulatory treatment of EAE with glatiramer acetate
Materials and Methods
(xxxvi) Animals. C57BL/6 mice were purchased from Harlan (Jerusalem,
Israel). Yellow fluorescent protein (YFP) 2.2 transgenic mice, (originated
from
C57BL/6 and CBA hybrids), which selectively express YFP on their motor and
sensory neuronal population (Feng et al., 2000), were kindly provided by J. R.
Sanes (Washington University St. Louis, MO). Female mice, 8-10 weeks of age,
were used in all experiments.
(xxxvii) EAE. Disease was induced by immunization with the peptide p35-
55 of rat MOG (SEQ ID NO:39), (Sigma, St. Louis, MO). Mice were injected
subcutaneously at the flank, with a 200 1 emulsion containing 300 g of MOG
in
CFA and 500 [tg of heat-inactivated Mycobacterium tuberculosis (Sigma). An
identical booster was given at the other flank one week later. Pertussis toxin

(Sigma), 300 [tg/mouse, was injected intravenously immediately after the first

MOG injection and 48 h later. Mice were examined daily. EAE was scored as
follows: 0, no disease; 1, limp tail; 2, hind limb paralysis; 3, paralysis of
all four
limbs; 4, moribund condition; 5, death.
(xxxviii) Glatiramer acetate (GA, CopaxoneS, Copolymer 1) consists of
acetate salts of synthetic polypeptides containing four amino acids: L-
alanine, L-
glutamate, L-lysine, and L-tyrosine. GA from batch 242990599, with an average
molecular weight of 7300 kDa, obtained from Teva Pharmaceutical Industries
86

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
(Petach Tikva, Israel) was used throughout the study. GA treatment was applied
by
5-8 consecutive daily subcutaneous injections (2 mg/mouse) in different stages
of
disease [i.e., (1) starting immediately after EAE induction (prevention
treatment),
(2) starting after appearance of disease manifestations at day 20 (suppression
treatment), or (3) starting during the chronic phase 45 days after induction
(delayed
suppression).
(xxxix) BrdU. 5-Bromo-2'-deoxyuridine (BrdU, Sigma), a thymidine analog
incorporating into the DNA of dividing cells, was injected intraperitoneally
(50
mg/kg), either concurrently with GA treatment (once each day), or immediately
after completion of GA injections (twice each day).
(xl) Perfusion. Animals were deeply anesthetized with Nembutal and
perfused transcardially with 2.5% paraformaldehyde. Brains were removed,
postfixed in 1% paraformaldehyde and cryoprotected with 15% sucrose solution
in
PBS. Free-floating sections (16 j.tm thick) were cut coronally or sagitaly
with a
sliding microtome (Leica SM 2000r) through the entire brain and collected
serially
in PBS.
(xli) Immunohistochemistry. To detect BrdU-incorporated cells, sections
were denatured in 2M HC1 in PBS at 37 C for 30 min and then neutralized with
0.1M borate buffer (pH 8.5) for 10 min at room temperature. To detect specific
cell
types, sections were pre-incubated in PBS solution containing 20% serum and
0.5%
Triton-X-100 for lh, and then incubated overnight at room temperature with
primary antibodies. The following primary antibodies were used: goat anti-
doublecortin (DCX) C-18 (1:200, Santa Cruz Biotechnology, Santa Cruz, CA),
mouse anti-NeuN (1:300, Chemicon, Temecula, CA), mouse anti-GFAP (1:100,
Pharmingen, San Jose, CA), rabbit anti-phospho-histone (1:200, Upstate
Biotechnology, Charlottesville, VA), rat anti-BrdU (1:200, Harlan,
Indianapolis,
IN), rat anti-CD1lb (1:50, Pharmingen) and chicken anti-BDNF (1:50, Promega,
Madison, WI). The second antibody step was performed by labeling with highly
cross-absorbed Cy2- or Cy3-conjugated antibodies to rat, mouse, rabbit, goat
or
chicken (Jackson ImmunoResearch, West Grove, PA), to avoid cross-reactivity,
87

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
(1:200; 20-40 min). Control slides were incubated with secondary antibody
alone.
In some cases, to enhance the signal, we used biotinylated secondary
antibodies for
90 min, followed by Cy2- or Cy3-conjugated Streptavidin (Jackson Immuno-
Research). Sections were stained with Hoechst 33258 (Molecular Probes) for
nuclear labeling. For detection of apoptosis, we used either rabbit anti-
cleaved
caspase-3 (1:75, Cell Signaling Technology, Beverly, MA) or TUNEL assay
(Apoptag Fluorescein Detection Kit, Intergen, Purchase, NY). In addition, we
used
Fluro-Jade B derivate (Chemicon), which specifically binds to degenerating
neurons.
(xlii) Microscopy. Stained sections were examined and photographed by a
confocal microscope (Axiovert 100M; Zeiss, Oberkochen, Germany), or by a
fluorescence microscope (E600; Nikon, Tokyo, Japan), equipped with Plan Fluor
objectives connected to CCD camera (DMX1200F, Nikon). Digital images were
collected and analyzed using Image Pro+software. Images were assembled using
Adobe Photoshop (Adobe Systems, San Jose, CA).
(xliii) Quantification. Neuronal progenitor cells were quantified by counting
the BrdU+ cells (those with BrdU/DCX dual staining) and by counting DCX +
cells
(in the SGZ) or by measuring the DCX stained area (in the SVZ and RMS, where
density did not permit counting of individual cells). Quantification was
performed
in coronal sections, in the SVZ starting at the level of the medial septum and
640
pm backward, and in the hippocampal DG (in both blades for BrdU/DCX or in the
upper blade for DCX) through its septotemporal axis. Quantification in the RMS

was done on sagital sections starting at 1 mm from the median line of the
brain and
640 ium laterally. The Results for each brain structure were averaged from 8
unilateral levels per mouse (80 lam apart, 3-4 mice in each treatment group)
and are
expressed as change fold from naive controls. Quantification of BrdU/NeuN
double
positive cells in the cortex was performed in areas of 0.15 mm2, selected at
random
(10 sections counted/mouse, 3 mice in each treatment group).
(xliv) Statistical analysis. For BrdU and DCX analysis, the mean SEM
(averaged from 8 unilateral levels per mouse, 3-4 mice in each treatment
group) was
88

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
subjected to one-way analysis of variance (ANOVA), followed by Fishers' least
significant difference (LSD) at comparison-wise error rate of 0.05, where
appropriate. Since control values for BrdU incorporation were reduced as a
function
of time, results were expressed as change fold from naive controls injected
concurrently with BrdU. The number of BrdU/NeuN double-positive cells in the
cortex was averaged from 10 sections per mouse (3 mice in each treatment
group)
and expressed as cells per mm3.
(xlv) Preparation of stem cells. ROSA26 mice express lacZ in all tissues of
the embryo and in most tissues of the adult mouse. Bone marrow (BM) cells were
isolated from ROSA26 mice by flushing the femur and tibias with Hanks balanced
salt containing 10% fetal bovine serum. A single cell suspension was prepared
for
transplantation.
Example 5(1). Description of the experimental model
To study the manifestations of EAE as well as GA treatment in the CNS, we
used the MOG 35-55 peptide-induced EAE model in two mice strains: the C57BL/6
susceptible strain and the YFP 2.2 transgenic mice, which selectively express
YFP
(yellow fluorescent protein) on their motor and sensory neuronal population,
and
thus provide a simple tool to follow axonal/neuronal damage (Feng et al.,
2000).
YFP 2.2 mice were susceptible to MOG-induced EAE similarly to C57BL/6 mice
(Fig. 19A). In both strains, EAE induction resulted in chronic (non-remitting)

disease, starting on days 16-20 (increasing in severity, reaching an average
score of
3 by day 20-24), and maintained in chronic phase, grade 2-2.5, until
perfusion. GA
treatment was applied by 5-8 daily injections in different stages: (1)
starting
immediately after disease induction (prevention treatment); (2) starting after
appearance of disease manifestations at day 20 (suppression treatment); or (3)

starting during the chronic phase, 45 days after induction (delayed
suppression). GA
ameliorated the clinical manifestations of EAE regardless of the stage in
which it
was administered (Fig. 19B). The beneficial effect was stable over time and
sustained until the mice were killed. The in situ manifestations in brains of
EAE-
89

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
inflicted mice (EAE mice) versus EAE-induced mice treated with GA (EAE+GA)
were studied in comparison to brains of naive mice (control).
Example 5(2). Characterization of neurological damage
In YFP 2.2 mice, YFP was expressed mainly by axons and dendrites. Partial
population in the cerebral cortex and the hippocampus expressed YFP in cell
bodies
as well. YFP expression in brain sections from mice that had suffered grade 2-
4
EAE revealed multiple neuronal malformations manifested in axonal transection,

sparse processes and fiber deterioration (Fig. 20A). Multiple widespread
lesions
were frequently observed in various brain regions (Fig. 20B), indicative of
considerable neuronal and axonal loss. An additional deformation in cell
morphology in EAE mice was enlargement and swelling of neuronal cell body
accompanied by margination of the nucleus as evident by distended hollow
;
Hoechst-stained nuclei (Fig. 20C). These defects did not result from
abnormality of
the transgenic strain, since similar phenomena were observed in EAE-induced
C57BL/6 mice stained by the neuronal marker NeuN (data not shown). Staining
with Fluoro-jade B, which binds to degenerating neurons, revealed positively
stained cells in the cortex, 25 days after disease induction, which is the
peak of
clinical manifestations (Fig. 20D). Yet, we could not see significant amount
of
apoptosis in the cortex and the striatum of both strains using either cleaved
caspase-
3 antibody or TUNEL assay, indicating that apoptotic mechanisms could not
account for the damage extent in this model. Perivascular infiltrations of CD3-

stained cells were found adjacent or inside aberrant regions, indicating the
detrimental role of infiltrating T-cells (Fig. 20A). In naive controls as well
as in
mice injected with GA but not induced with EAE, we did not find neuronal
malformations or perivascular infiltrations (not shown).
In brains of EAE+GA mice (either prevention or suppression treatment),
considerably less damage was detected than in brains of EAE mice, revealing a
smaller amount of deteriorating fibers (Fig. 20A), reduced number and size of
lesions (Fig. 2B) and less swollen cell nuclei (Fig. 20C). A thin layer of YFP

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
positive fibers was frequently found over the lesions in the GA treated
animals (Fig.
B), suggesting surviving filaments or axonal sprouting in the damaged areas. T-
cells
infiltrations were found also in brains of GA treated mice, yet, in smaller
amount
and their position was not associated with damage (Fig. 20A).
Example 5(3). Microglia activation
Immunostaining for MAC-1 (CD11b, expressed on macrophages and
microglia and up-regulated after their activation), correlated with the extent
of
neuronal injury in EAE mice (shown in the cerebellum, Fig. 21A). Thus, in
areas
occupied with activated microglia, sparse fibers and axonal loss were
generally
evident (box I), whereas in adjacent areas of non-activated microglia,
neuronal
structure seemed intact (box II). Perivascular infiltration of activated MAC-
1+ cells
was found in injured areas suggesting that peripherally originated macrophages

were also involved in the pathological process. As shown in Fig. 21B, the vast
increase in MAC-1 staining intensity found in EAE mice was demonstrated in
additional brain regions e.g. striatum, thalamus and hippocampus. MAC-1+ cells
in
brains of control mice had relatively small cell body and long branched
processes
indicative of resting microglia. In contrast, brains of EAE mice, manifested
rounding cell body with increased size and numerous retracted short processes,
indicative of highly activated microglia (insert). In brains of EAE+GA mice,
MAC-
1 expression was significantly reduced, exhibiting moderate extent of
activation.
Cell morphology of MAC-1+ cells in GA-treated mice was similar to that of non-
activated microglia in naive mice (insert). This arrest of microglial
activation in
EAE+GA mice was found at various times up to 30 days after termination of GA
injections.
Example 5(4). Proliferation of neuronal progenitor cells
To evaluate the generation and proliferation of neuronal progenitor cells
following the pathological process of EAE, as well as after GA treatment, we
used
two markers: the immature neuronal marker DCX (associated with migrating and
91

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
differentiating neurons of fetal and adult brain), and BrdU (thymidine analog
incorporating into DNA of dividing cells) that had been injected concurrently
with
GA treatment. Hence, DCX expression indicated the amount of new neurons
generated 10-14 days before animal sacrifice, and the number of BrdU
incorporated
cells (those with BrdU/DCX dual staining) indicated the number of
neuroprogenitors emerging during the BrdU injection period. Neuroproliferation

was studied in the neuroproliferative zones ¨ the subventricular zone (SVZ) as
well
as in the subgranular zone (SGZ) and the granular cell layer (GCL) of the
hippocampus. BrdU and DCX manifested overlapping patterns.
In the SVZ of EAE mice, neuroprogenitor proliferation was elevated
following disease appearance (25 days after EAE induction, 1 day after last
BrdU
injection) in comparison to the controls (Figs. 22A, 22BI). This was evident
by a
2.1 and a 1.4 fold increase in BrdU and DCX expression, respectively (Fig.
22D,
SVZ, I, red columns). Still, 10 and 20 days later there was no significant
difference
in BrdU and DCX expression between EAE mice and naïve controls (Figs. 2211,
22D, II, III). Furthermore, in mice enduring disease for prolonged periods (35
and
60 days), proliferation manifested by BrdDU incorporation was lower than that
of
controls, either when BrdU was injected concurrently with EAE induction and
tested one month later (Fig. 22D, first red column) or during the chronic
stage
before perfusion (Fig. 22D, last red column). GA treatment in EAE mice
(administration schedules illustrated in Fig. 22E) augmented neuronal
proliferation
in the SVZ in comparison to untreated EAE mice, as well as to controls (Figs.
22A,
22B). This elevation reached statistical significance over control and EAE for
both
BrdU and DCX by the suppression treatment, 1 and 10 days after termination of
GA
injection (Fig. 22D). Delayed suppression treatment resulted in significant
elevation
over EAE but not control. In the prevention treatment, substantial elevation
over
EAE and control was observed only for DCX (4-fold), though even BrdU
incorporation was indicative of significant elevation compared to EAE.
In the SGZ of the hippocampus, neuronal proliferation was elevated
following disease appearance, but subsequently declined below that of naive
control
92

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
(Figs. 22C, 22D, hippocampus, red columns). The effect of GA treatment in the
hippocampus was similar to its effect in the SVZ, namely increased
proliferation
manifested by both BrdU incorporation and DCX expression that was not
sustained
after termination of the suppression treatments. Prevention as well as delayed
suppression treatments resulted in higher neuroproliferation than in EAE mice,
one
month and one day after termination of GA/BrdU injection, respectively.
Notably,
in the hippocampus of EAE mice, and to a greater extent in EAE+GA mice (Fig.
22C), BrdU/DCX+ cells were found in the SGZ and in the adjacent GCL. The
DCX expressing cells manifested dense and branched dendritic tree with well-
developed apical dendrites that crossed the inner molecular layer and extended
into
the outer molecular layer.
GA injection to naive mice (without EAE), either just prior to perfusion or a
month earlier, did not result in significant elevation of BrdU or DCX
expression, in
both the SVZ and the DG (Fig. 22D, gray columns).
Example 5(5). Migration of neuronal progenitor cells
To study the destiny of the induced progenitor cells, we first followed their
mobilization into the route in which SVZ cells normally migrate in adult mice -
the
rostral migratory stream (RMS, illustrated in Fig. 23A). As depicted in a
segment
adjacent to the SVZ (Fig. 23D) and in a more medial section (Fig. 23E), the
amount
of BrdU as well as DCX labeled cells migrating along the RMS of EAE mice (25
days post disease induction, 1 day after last BrdU injection), was elevated in

comparison to controls. This was manifested by a 3.1 and a 1.6 fold elevation
in the
number of BrdU/DCX cells and in the DCX stained area, respectively (Figs. 23F,
23G, red columns). After GA treatment (on days 20-25, suppression), the amount
of
neuronal progenitors in the RMS was even higher, exhibiting an extensive
stream of
BrdU/DCX expressing cells (Figs. 23B, 23D, 23E). Thus, an increase of 7.8 and
2.6
fold in BrdU and DCX expression over control, and 2.2 and 1.6 fold over EAE
mice
was obtained following GA treatment (Figs. 23F, 23G, blue columns). Notably,
93

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
injection of GA alone also enhanced mobilization of progenitors into the RMS,
but
to a lower extent (Figs. 23F, 23G, gray columns).
Similar mobilization patterns of neuronal progenitors were found at a later
time point (35 days after EAE induction, when GA treatment was given as
prevention treatment, i.e., elevated DCX expression in the RMS of EAE vs.
control
mice, and even more robust migration in EAE+GA mice) (Fig. 23H).
Interestingly,
in one EAE mouse (out of 13 mice), we found enhanced neuronal migration
similar
to that of GA-treated mice. This mouse (denoted EAE-rec) exhibited only
slight,
short term disease (score 2, at days 24-26 after induction), and completely
recovered by the day of perfusion.
Treatment of EAE mice with GA led not only to enhanced mobilization of
neuronal progenitors through the RMS, but also to their migration into a
region
corresponding to the lateral cortical stream (LCS) of neuronal migration,
naturally
found in the developing forebrain (illustrated in Fig. 23A). Hence, DCX+ cells
appeared to travel from the SVZ caudally, in a chain along the corpus callosum
and
the hippocampo-callosal interface, towards various cortical regions mainly the

occipital cortex (Fig. 23C). We could not trace such mobilization patterns in
corresponding sections of EAE mice not treated with GA. Furthermore, in
EAE+GA mice, neuronal progenitors diverged from the classic neuroproliferative
zones, as well as the migratory streams and spread to atypical regions such as
the
striatum, nucleus accumbens and the cortex (Fig. 24). The DCX+ cells appeared
to
move away from the RMS in close proximity to YFP expressing filaments,
suggesting their migration along nerve fibers (Fig. 24A). As seen by their
direction
and orientation, they migrated away from both the RMS and the SVZ, yet in some
mice most cells extended from the SVZ (Fig. 24B), whereas in others the RMS
seemed as their major origin (Fig. 24C). DCX+ cells appeared to reach into the

frontal cortex from the RMS (Figs. 23B, 24D) and to the occipital cortex from
the
LCS (Fig. 23C). They manifested morphological features characteristic of
migrating neurons, such as fusiform somata with a leading and trailing process
(O'Rourke et al., 1995), and their orientation was consistent with migration
away
94

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
from the migratory stream into the internal part of the cortex, along nerve
fibers
(shown in layers 5 and 6, Figs. 24E, 24F). We did not detect neuroprogenitors
in
areas remote from the neuroproliferative zones and the migratory streams such
as
the cerebellum and the pons.
At early time point following GA treatment and BrdU injection (1-10 days),
neuronal progenitors that migrated away from the RMS manifested BrdU and DCX
co-expression as shown in the striatum (Fig. 25A) and the accombens nucleus
(Fig.
25B), indicating that they underwent division concurrently with GA treatment.
In
some cases, these double positive cells appeared in small clusters, suggesting
local
divisions as well. Furthermore, staining with phosphorylated histone H3, an
endogenous marker of cells in M phase, indicated that some DCX+ cells had
proliferated just prior to perfusion, as seen for the neuroprogenitors
accumulated in
the nucleus accombens in Fig. 25C.
At later time points (one month after completion of GA treatment), BrdU+
cells co-expressing the neuronal nuclear antigen (NeuN), were found in the
striatum
(Figs. 25D, 25F), nucleus accumbens (Fig. 25E), and cortex (Fig. 25G,
cingulate
cortex layer 5), indicating that some neuroprogenitor cells have
differentiated
further toward a mature neuronal phenotype. In the cortex (Fig. 25H, 251,
cingulate,
Fig. 25J, occipital, Fig. 25K, motor) of YFP mice, pyramidal cells co-
expressing
BrdU and YFP with apical dendrites and axons were observed, indicative of
mature
functional neurons. An average of 128 + 46/mm3 BrdU+/NeuN+ double-labeled
cells
were found in the cortex of EAE+GA mice, consisting of 1.3% of all NeuN+
cells. It
should be noted that BrdU+/NeuN+ cells were found also in the cortex of EAE
mice
not treated with GA, though fewer (48 + 25/mm3, 0.58% from NeuN+ cells). In
the
cortex of naive mice, BrdU+/NeuN+ cells were not found.
Example 5(6). Migration to lesion sites
The newly generated neurons seemed to be attracted to damaged regions.
Hence, clusters of DCX/BrdU as well as NeuN/BrdU co-expressing cells, were
situated in areas with deteriorating YFP expressing fibers and lesions (Figs.
24B,

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
24C, 25A-25D). Furthermore, DCX expressing cells, were found around the
margins and inside lesions in the striatum (Figs. 26B, 26C), cortex (Figs.
26D, 26E)
and the nucleus accombens (Fig. 26F). In EAE mice (not treated by GA), a few
DCX+ cells surrounding lesions also observed (Fig. 26A), but in EAE+GA mice
the
amount of progenitors migrating to the lesions was much higher. The DCX+
neuroprogenitors localized into areas extensively occupied with astrocytes
expressing GFAP (Figs. 27A-27C), suggesting their migration into gliotic scar
areas.
In lesions occupied by DCX+ cells (Figs. 26D-26F), we observed YFP
expressing fibers extending into lesions, suggesting the induction of axonal
regeneration, or sprouting, by the neuroprogenitors. To find out if the newly
generated neurons can actually induce a growth-promoting environment, we
tested
their ability to express BDNF. As shown in Fig. 27, in the nucleolus accumbens

(Figs. 27D, 27E), and the hippocampus (Fig. 27F), substantial proportion of
the
migrating DCX+ cells, in EAE+GA mice, manifested extensive expression of
BDNF.
Example 5 (7). Combination treatment of glatiramer acetate and progenitor
stem cell in a mice model of EAE.
The following experiment was carried out in order to assess the effect of the
combination of glatiramer acetate (GA) and stem cells administration in an EAE

model. We employed the myelin oligodendrocyte glycoprotein (MOG) induced
EAE mice model. Neuronal and axonal degeneration are extensively manifested in

EAE mice. GA treatment was applied in EAE mice in combination with stem cells
by a procedure, which was found to be effective in generating self-
neurogenesis,
namely by daily subcutaneous injections.
Multipotent stem cells were obtained from bone marrow of ROSA26
transgenic mice, which express Lac-z in most tissues of the adult mouse.
Expression
of Lac-z gene was detected by enzymatic activity of the gene product beta-
galactosidase. These stem cells were transplanted into EAE C57BL/6 mice by
local
96

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
stereotactical inclusion into the lateral ventricle of the brain.
Alternatively, it is
possible to administer stem cells systemically by intravenous injection.
The effect of combined GA and stem cells administration was compared to
administration of GA and stem cells separately. Untreated EAE mice and naive
mice served as controls. After treatment, mice were inspected daily for
neuronal
symptoms and scored for disease severity and/or clinical improvement. The in
situ
effect of the treatments was evaluated by characterization of neuronal damage
as
described in 5(2) above. The fate of the transplanted cells was monitored
using
immunohistological methods as described in 5(4), 5(5) and 5(6) above. For
example, proliferation was assessed by using markers such as BrdU injected
concurrently with transplantation and differentiation was monitored by
detecting
DCX and NeuN markers. We also followed the migration of the transplanted cells

and their ability to reach the lesion site.
Preliminary results obtained indicate that combined GA+stem cell treatment
augmented the beneficial effect of each treatment separately, as evidenced by
the
above parameters inspected. The same beneficial effects can be obtained by the

combined treatment of GA and stein cells in other experimental models. Thus,
the
combined treatment of GA and stem cells can be used in therapy of additional
neurological diseases and other disorders.
Discussion
The major finding reported here is that peripheral immunomodulatory
treatment of an inflammatory autoimmune neurodegenerative disease induces
neuroprotection as well as augmentation of the self-neurogenesis triggered by
the
pathological process. This results in massive migration of new neurons into
injury
sites, in brain regions that do not normally undergo neurogenesis, suggesting
relevance to the beneficial effect of GA in EAE and MS.
The histopathological manifestations of MOG-induced EAE, in both
C57BL/6 and YFP 2.2 strains were deteriorating fibers, axonal loss, widespread

lesions, and nucleus margination, indicative of severe damage (Fig. 20).
97

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Perivascular infiltrations of T-cells (Fig. 20A) and macrophages (Fig. 21A)
were
found in close proximity to aberrant regions, in consistence with their
detrimental
role in this disease (Behi et al., 2005; Stollg and Jander 1999). The
protective effect
of GA was manifested in prevention of the typical axonal and neuronal damage
as
evidenced in less deteriorating fibers, reduced amount of lesions with smaller
magnitude and less marginized cell nuclei. Additional prominent effect of GA
was
the reduction in microglia activation (Fig. 21B), manifested in all time
points (1-30
days after treatment termination), by the various schedules. Microglia
function as
antigen-presenting cells within the CNS and thereby activate encphalitogenic T-

cells and produce inflammatory toxic mediators, though, dual function due to
their
capacity to express neurotrophic factors was also demonstrated (Stollg and
Jander
1999). In the current model, microglia activation was markedly elevated in EAE

inflicted mice in various brain regions, and this activation correlated with
the
amount of neuronal injury.
GA treatment resulted not only in decreased neuronal damage but also in
increased neuronal proliferation. The combination of two detection markers
allowed
us to evaluate both the amount of new neurons generated 10-14 days before the
animal was killed, by the overall expression of the immature neuronal marker
DCX
(Bayer et al., 1991) as well as the number of neuroprogenitors emerging during
the
concurrent BrdU/GA injection period (those that differentiated into the
neuronal
lineage and thus presented BrdU/DCX dual staining). Both systems gave
comparable results as to the effect of the pathological process of EAE and
that of
GA treatment. Hence, EAE induction triggered increased neuroprogenitor
proliferation in the neuroproliferative zones (the SVZ and the SGZ) following
disease appearance (Fig. 22), in accordance with previous studies
demonstrating
increased cell proliferation in these zones following injury (Jin et al.,
2003; Magavi
et al., 2000; Picard-Riera et al., 2002). Still this neuroproliferation
decreased
gradually and subsequently declined below that of naive mice, indicative of
the
impairment inflicted by the disease and the failure of self-neurogenesis to
compensate for the damage. GA treatment applied by various schedules to EAE
98

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
mice augmented neuronal proliferation in both the SVZ and the SGZ over that of

EAE mice and prolonged its duration. Of special significance is the
neuroproliferative consequence of GA treatment initiated in the chronic phase
of the
disease (delayed suppression), as this phase in EAE/MS is regarded as the
stage in
which exhausted self-compensating neurogenesis fails, and extensive
neurodegeneration overcomes (Bjartmar et al., 2003; Hobom et al., 2004).
Neuroprogenitors originated in the SVZ were mobilized into the route in
which they normally migrate in adults, the RMS. This mobilization was
increased in
EAE mice, and GA augmented it even further (Fig. 23). The therapeutic
relevance
of this effect is implied by the enhanced neuronal migration found in the EAE
mouse that exhibited slight, short-term disease and spontaneous recovery.
Still, in
GA treated mice neuroprogenitor, migration was not confined to the RMS. We
found recurrence of the LCS - neuronal migratory route, naturally found in the

embryonic forebrain (Francic et al., 1999), as DCX-expressing cells migrated
along
the corpus callosum and the hippocampo-callosal interface, towards various
cortical
regions mainly to the occipital cortex (Fig. 23C). Furthermore, neuronal
progenitors
diverged from the classic neuroproliferative zones, as well as the migratory
streams
and spread to adjacent atypical brain regions that do not normally undergo
neurogenesis such as the striatum, nucleus accumbens and the cortex (Fig. 24).
In
the hippocampus of EAE mice, subsequent to disease appearance and to a greater
extent and longer duration in EAE+GA mice, BrdU and DCX expressing cells
migrated from the SGZ into the adjacent GCL, extending branched dendrites
through the inner and outer molecular layer (Fig. 22C). However, mobilization
of
SGL originating cells was probably restricted to the hippocampus, as we found
no
evidence for migration beyond this region, in accord with previous studies
identifying the SVZ rather than the SGZ as the source of neuroprecursor
migration
(Jin et al., 2003).
At early time points following GA and BrdU injection (1-10 days after their
last injection), BrdU + neuroprogenitors expressed the immature neuronal
marker
DCX characteristic to migrating and differentiating neurons (Bernier et al.,
2002),
99

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
and displayed migratory morphology, fusiform somata with a leading and a
trailing
process (O'Rouke et al., 1995) (Fig. 24). It has been doubted whether
progenitors
retain their ability to proliferate after leaving the neuroproliferative zones
(Gould
and Gross, 2002; Iwai et al., 2002). In EAE+GA mice, we found small clusters
of
BrdU/DCX co-expressing cells in the striatum and the nucleus accumbens,
suggesting local divisions. Furthermore, staining with phosphorylated histone
an
endogenous marker of cells in M phase indicated that some DCX+ cells in these
regions had divided just prior to perfusion (Fig. 25), suggesting in situ
proliferation
outside the classic neuroproliferative zones. At later time point (one month
after
completion of GA treatment), DCX+ cells with branching processes (Fig. 26) as
well as BrdU+ cells expressing the mature neuronal marker NeuN and displaying
mature morphology were observed (Fig. 25). The amount of new neurons in the
cortex of EAE mice was comparable to those found in other cases of damage-
induced neurogenesis (Magavi et al., 2000; Picard-Riera et al., 2002; Arlotta
et al.,
2003). GA treatment increased this number by 2.6 fold, indicating substantial
elevation of newly generated neurons. BrdU/NeuN+ cells were not found in
cortex
of naive mice, confirming that neurogenesis does not normally occur in the
adult
rodent cortex (Iwai et al., 2002; Jin et al., 2003; Arlotta et al., 2003).
Thus, three
processes comprising neurogenesis: cell proliferation, migration and
differentiation
(Jin et al., 2003; Chen et al., 2004), were elevated after GA treatment.
These findings establish correlation between GA treatment and generation of
neuroprotection and neurogenesis. It is possible that these effects result
from
suppression of inflammation - the insult initiating the pathological process,
and thus
the subsequent damage, as demonstrated for anti-inflammatory treatment after
endotoxin administration (Monje et al., 2003). The ability of GA to shift
cytokine
secretion from the Thl inflammatory to the Th2/3 anti-inflammatory pathway was

demonstrated in the periphery of mice and humans (Aharoni et al., 1998; Duda
et
al., 2000). Moreover, GA induces specific Th2/3-cells that cross the BBB,
accumulate in the CNS (Aharoni et al., 2000, 2002) and express in situ the
anti-
inflammatory cytokines IL-10 and TGF-I3 (Aharoni et al., 2003). In the present
100

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
study as well, infiltrating T-cells were found in brains of EAE+GA mice, and
in
contrast to EAE mice, their location was not associated with damage (Fig.
19C).
However, the effect of the GA-induced cells in the CNS goes beyond blockage of

inflammation. Hence, IL-10 was shown to modulate glial activation (Ledeboer
et.
al., 2000), thus its in situ expression may account for the blockade of
microglia
activation in GA treated mice (Fig. 21). As for TGF-P, its neuroprotective
activity
has been shown in various species (Dhandapani and Brann, 2003) as well as its
ability to induce neurproliferation and differentiation (Newmann eyal., 2000;
Kawauchi et al., 2003). Furthermore, GA-specific cells in the brain were shown
to
express the potent neurotrophic factor BDNF (Aharoni et al., 2003), a key
regulator
of neuronal survival and neurogenesis in the adult brain (Lassmann et al.,
2003).
BDNF was shown to stimulate recruitment of SVZ cells, their migration through
the
RMS to structures, which do not exhibit neurogenesis in adulthood, and their
differentiation into neurons (Pencea et al., 2001), similarly to the finding
in this
study. Of special relevance therefore are our previous finding that adoptive
transfer
of GA specific T-cells or GA injection as such, induced bystander effect on
CNS
resident cells e.g. astrocytes and neurons, to extensively express IL-10, TGF-
P and
BDNF, resulting in their significant elevation in various brain regions
(Aharoni et
al., 2003, 2004).
90 It
is of special significance that the newly generated neurons were attracted
or recruited to damaged regions, as evidenced by their migration into gliotic
scarred
areas (Fig. 27) and to regions exhibiting fiber deterioration, neuronal loss
and
lesions (Figs. 24B, 24C, 25A-25D, 26). Directed migration of new neurons
towards
injury sites has been demonstrated following cerebral ischemia (Jin et al.,
2003), as
well as in this study in EAE mice (Fig. 26A). However, although lesions in EAE
mice treated by GA were less extensive, the amount of progenitors migrating
into
them was drastically larger. These new neurons could constitute a pool for the

replacement of dead or dysfunctional cells and/or induce growth-promoting
environment that supports neuroprotection and axonal growth. The latter
activity
was evidenced by BDNF expression of the new neurons (Figs. 27D-27F).
101

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Moreover, in lesions occupied by neuroprogenitors, YFP expressing fibers
extending into the lesions were observed (Figs. 20B, 26D-26F), suggesting the
induction of axonal regeneration, or sprouting. The cumulative results
presented
here support the notion that an immunomodulatory drug can induce
neuroprotection
and neurogenesis that counteract the neurodegenerative disease course.
1 02

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
REFERENCES
Aberg, M. A., Aberg, N. D., Hedbacker, H., Oscarsson, J. & Eriksson, P. S.
Peripheral infusion of IGF-I selectively induces neurogenesis in the adult rat

hippocampus. J Neurosci 20, 2896-903 (2000).
Aharoni R, Schlegel PG, Teitelbaum D, Roikhel-Karpov 0, Chen Y, Arnon
R, Sela M, Chao NJ. (1997) Studies on the mehanism and specificity of the
effect of
synthetic random copolymer GLAT on graft-versus-host disease. Immunol. Lett.
58
(2):79-87.
Aharoni R, Teitelbaum D, Sela M, Arnon R (1998) Bystander suppression of
experimental autoimmune encephalomyelitis by T cell lines and clones of the
Th2
type induced by copolymer 1. J. Neuroimmunol. 2:135-146
Aharoni R, Teitelbaum D, Leitner 0, Meshorer A, Sela M, Arnon R (2000)
Specific Th2 cells accumulate in the central nervous system of mice protected
against experimental autoimmune encephalomyelitis by copolymer 1. Proc. Natl.
Acad. Sci. USA. 97:11472-11477.
Aharoni R., Teitelbaum D., Amon R., Sela M. (2001). Copolymer 1 inhibits
manifestations of graft rejection. Transplantation 27: 598-605.
Aharoni R, Meshorer A, Sela M, Arnon R (2002) Oral treatment of mice
with copolymer 1 (glatiramer acetate) results in the accumulation of specific
Th2
cells in the central nervous system. J Neuroirnmunol. 126:58-68.
Aharoni R, Kayhan B, Eilam R, Sela M, Arnon R (2003) Glatiramer acetate-
specific T cells in the brain express T helper 2/3 cytokines and brain-derived

neurotrophic factor in situ. Proc. Nat. Acad. Sci. USA. 100:14157-14162.
Aharoni R, Eilam R, Labunskay G, Sela M Arnon R (2004) Copaxone
(glatiramer acetate) injection augment brain derived neurotrophic factor
expression
in the brain. Multiple Sclerosis 10:S256
Aharoni R., Yussim A., Sela M., Arnon R. (2005). Combined treatment of
glatiamer acetate and low doses of immunosuppressive drugs is effective in the

prevention of graft rejection. Int Immunopharmacol. 5(1):23-32.
103

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Aharoni R, Kayhan B, Amon R (2005) Therapeutic effect of the
immunomodulator glatiramer acetate on trinitrobenzene sulfonic acid-induced
experimental colitis. Inflamm Bowel Dis. 11(2):106-115.
Angelov DN, Waibel S, Guntinas-Lichius 0, Lenzen M, Neiss WF, Tomov
TL, Yoles E, Kipnis J, Schori H, Reuter A, Ludolph A, Schwartz M. (2003).
Therapeutic vaccine for acute and chronic motor neuron diseases: implications
for
amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. U.S.A. 100(8):4790-4795.

Amon, R, Sela M. (2003) Immunomodulation by the copolymer glatiramer
acetate. J. Mol. Recognit. 16:412-421.
Bakalash S, Kessler A, Mizrahi T, Nussenblatt R, Schwartz, M. (2003)
Antigenic specificity of immunoprotective therapeutic vaccination for
glaucoma.
Invest. Ophthalmol. Vis. Sci. 44: 3374-3381.
Bayer SA, Altman J, Russo RJ, Dai XF, Simmons JA (1991) Cell migration
in the embryonic neocortex. I. Comp. Neurol. 307:499-516.
Behi 1\4E, Dubucquoi S, Lefranc D, Zephir H, De Seze J, Vermersch P, Prin
L (2005) New insights into cell responses involved in experimental autoimmune
encephalomyelitis and multiple sclerosis. Immunology Letters 96:11-26.
Benner EJ, Mosley RL, Destache CJ, Lewis TB, Jackson-Lewis V, Gorantla
S, Nemachek C, Green SR, Przedborski S, Gendelman HE. (2004) Therapeutic
immunization protects dopaminergic neurons in a mouse model of Parkinson's
disease. Proc Natl Acad Sci U S A. 101:9435-40.
Bernier PJ, Bedard A, Vinet J, Levesque M, Parent A (2002) Newly
generated neurons in the amygdala and adjoining cortex of adult primates.
Proc.
Nat. Acad. Sci. U S A. 99:11464-11469.
Bjartmar C, Wujek JR, Trapp BD (2003) Axonal loss in the pathology of
MS: consequences for understanding the progressive phase of the disease. J.
Neurol.
Sci. 15:165-171
Bliss, T.V. & Collingridge, G.L. A synaptic model of memory: long-term
potentiation in the hippocampus. Nature 361, 31-9 (1993).
104

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Boje, K. M. & Arora, P. K. Microglial-produced nitric oxide and reactive
nitrogen oxides mediate neuronal cell death. Brain Res. 587, 250-6 (1992).
Borchelt, DR et al., Neuron 19, 939 (1997).
Brown, J. et al. Enriched environment and physical activity stimulate
hippocampal but not olfactory bulb neurogenesis. Eur J Neurosci 17, 2042-6
(2003).
Brown JP, Couillard-Despres S, Cooper-Kuhn CM, Winkler J, Aigner L,
Kuhn HG. (2003) Transient expression of doublecortin during adult
neurogenesis. J.
Comp Neurol. 467: 1-10.
Butovsky 0, Talpalar AE, Ben-Yaakov K, Schwartz M. Activation of
microglia by aggregated beta-amyloid or lipopolysaccharide impairs MHC-II
expression and renders them cytotoxic whereas IFN-gamma and IL-4 render them
protective. Mol Cell Neurosci 29:381-393 (2005).
Butovsky, O., Hauben, E. & Schwartz, M. Morphological aspects of spinal
cord autoimmune neuroprotection: colocalization of T cells with B7--2 (CD86)
and
prevention of cyst formation. Faseb J. 15, 1065-7 (2001).
Cameron, H.A. & McKay, R.D. Restoring production of hippocampal
neurons in old age. Nat Neurosci 2, 894-7 (1999).
Cameron, H.A. & McKay, R.D. Adult neurogenesis produces a large pool of
new granule cells in the dentate gyrus. J Comp Neurol 435, 406-17 (2001).
Cammer, W. & Zhang, H. Maturation of oligodendrocytes is more sensitive
to TNF alpha than is survival of precursors and immature oligodendrocytes. J
Neuroimmunol 97, 37-42 (1999).
Chen J, Magavi SS, Macklis JD (2004) Neurogenesis od corticospinal motor
neurons extending spinal projections in adult mice. Proc. Nat. Acad. Sci. USA.
101:16357-16362.
Cummings, B. J., Uchida, N., Tamaki, S. J., Salazar, D. L., Hooshmand, M.,
Summers, R., Gage, F. H. & Anderson, A. J. Proc Natl Acad Sci USA 102, 14069-
74 (2005).
105

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Dawson, M. R., Polito, A., Levine, J. M. & Reynolds, R. NG2-expressing
glial progenitor cells: an abundant and widespread population of cycling cells
in the
adult rat CNS. Mol Cell Neurosci 24, 476-88 (2003).
Dhandapani KM, Brann DW (2003) Transforming growth factor-beta: a
neuroprotective factor in cerebral ischemia. Cell Biochem Biophs. 39:13-22.
Dijkstra, C. D., De Groot, C. J. & Huitinga, I. The role of macrophages in
demyelination. J Neuroimmunol 40, 183-8 (1992).
Duda, P.W., Schmied, M.C., Cook, S.L., Krieger, J.I., and Hafler, D.A.
Glatiramer acetate (Copaxone) induces degenerate, Th2-polarized immune
responses in patients with multiple sclerosis. J Clin Invest 105:967-976
(2000).
Ekdahl, C. T., Claasen, J. H., Bonde, S., Kokaia, Z. & Lindvall, O.
Inflammation is detrimental for neurogenesis in adult brain. Proc Natl Acad
Sci U S
A100, 13632-7 (2003).
Engesser-Cesar, C., Anderson, A.J., Basso, D.M., Edgerton, V.R. & Cotman,
C.W. Voluntary wheel running improves recovery from a moderate spinal cord
injury. J Neurotrauma 22, 157-71 (2005).
Enzmann, G. U., Benton, R. L., Woock, J. P., Howard, R. M., Tsoulfas, P. &
Whittemore, S. R. Exp Neurol 195, 293-304 (2005).
Feng G, Mellor RH, Bernstein M, Keller-Peak Cynthia, Nguyen QT, Wallace M,
Nerbonne JM, Lichtman JW, Samce JR (2000) Imaging neuronal subsets in
transgenic mice expressing multiple spectral variants of GFP. Neuron 28:41-51.

Francis F, Koulakoff A, Boucher D, Chafey P, Schaar B, Vinet MC,
Friocourt G, McDonnell N, Reiner 0, Kahn A, McConnell SK, Berwald-Netter Y,
Denoulet P, Chelly J. (1999) Doublecortin is a developmentally regulated
microtobule-associated protein expressed in migrating and differentiating
neurons.
Neuron 23:247-256.
Fridkis-Hareli M, Aharoni R, Teitelbaum D, Arnon R, Sela M, Strominger
JL. (1999) Binding motifs of copolymer 1 to multiple sclerosis- and rheumatoid

arthritis-associated HLA-DR molecules. J Immunol. 162(8):4697-4704.
106

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Gould E, Gross CG (2002) Neurogenesis in adult mammals: some progress
and problems. J. Neuroscience. 22:619-623.
Gould, E., Beylin, A., Tanapat, P., Reeves, A. & Shors, T.J. Learning
enhances adult neurogenesis in the hippocampal formation. Nat Neurosci 2, 260-
5
(1999).
Hauben E, Schwartz M. Therapeutic vaccination for spinal cord injury:
helping the body to cure itself. Trends Pharmacol Sci 24:7-12 (2003).
Hauben, E., Gothilf, A., Cohen, A., Butovsky, O., Nevo, U., Smimov, I.,
Yoles, E., Akselrod, S. & Schwartz, M. (2003) J Neurosci 23, 8808-19.
Hauben, E., Agranov, E., Gothilf, A., Nevo, U., Cohen, A., Smimov, I.,
Steinman, L. & Schwartz, M. Posttraumatic therapeutic vaccination with
modified
myelin self-antigen prevents complete paralysis while avoiding autoimmune
disease. J Clin. Invest. 108, 591-9 (2001).
Hauben, E., Butovsky, O., Nevo, U., Yoles, E., Moalem, G., Agranov, G.,
Mor, F., Leibowitz-Amit, R., Pevsner, E., Akselrod, S., Neeman, M., Cohen, I.
R. &
Schwartz, M. Passive or active immunization with myelin basic protein promotes

recovery from spinal cord contusion. J. Neurosci. 20, 6421-6430 (2000).
Hellings N, Raus, J. Stinissen P (2002) Insights into the
immunopathogenesis of multiple sclerosis. Immunol Res. 25:27-51.
Hobom M, Storch MK, Weissert R, Maier K, Radhakrishnan A, Kramer B,
Bahr M, Diem R. (2004) Mechanisms and time course of neuronal degeneration in
experimental autoimmune encephalomyelitis. Brain Pathol. 14:148-157.
Hsieh, J., Aimone, J.B., Kaspar, B.K., Kuwabara, T., Nakashima, K., and
Gage, F.H. 2004. IGF-I instructs multipotent adult neural progenitor cells to
become oligodendrocytes. J Cell Biol 164, 111-22 (2004).
Imitola, J., Raddassi, K., Park, K. I., Mueller, F. J., Nieto, M., Teng, Y.
D.,
Frenkel, D., Li, J., Sidman, R. L., Walsh, C. A., Snyder, E. Y. & Khoury, S.
J. Proc
Natl Acad Sci USA 101, 18117-22 (2004a).
Imitola, J., Comabella, M., Chandraker, A. K., Dangond, F., Sayegh, M. H.,
Snyder, E. Y. & Khoury, S. J. Neural stem/progenitor cells express
costimulatory
107

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
molecules that are differentially regulated by inflammatory and apoptotic
stimuli.
Am J Pathol 164, 1615-25 (2004b).
Ishii, K. et al., Faseb .114, 1485 ( 2000).
Iwai M, Sato K, Omori N, Nagano I, Manabe Y, Shoji M, Abe K. (2002)
Three steps of neural stem cells development in gerbil dentate gurus after
transient
ischemia J. Cereb. Blood Flow Metab. 22:411-419.
Jankowsky, JL et al., Hum Mol Genet 13, 159 (2004).
Jin K, Sun Y, Xie L. Peel A, Mau X0, Batteur S, Greenberg DA (2003)
Directed migration of neuronal precureors into the ischemic cerebral cortex
and
striatum. Molecular and Cellular Neuroscience 24:171-189.
Jones, L. L., Yamaguchi, Y., Stallcup, W. B. & Tuszynski, M. H. NG2 is a
major chondroitin sulfate proteoglycan produced after spinal cord injury and
is
expressed by macrophages and oligodendrocyte progenitors. .1 Neurosci 22, 2792-

803 (2002).
Kawauchi S, Beites CL, Crocker CE, Wu HH, Bonnin A, Murray R, Calof
AL. (2004) Molecular signals regulating proliferation of stern and progenitor
cells
in mouse olfactory epithelium. Dev. Neurosci. 26:166-180.
Kempermann, G., Jessberger, S., Steiner, B. & Kronenberg, G. Milestones of
neuronal development in the adult hippocampus. Trends Neurosci 27, 447-52
(2004).
Kempermann, G., Kuhn, H.G. & Gage, F.H. More hippocampal neurons in
adult mice living in an enriched environment. Nature 386, 493-5 (1997).
Kipnis, J., Cohen, H., Cardon, M., Ziv, Y. & Schwartz, M. T cell deficiency
leads to cognitive dysfunction: Implications for therapeutic vaccination for
schizophrenia and other psychiatric conditions. Proc. Natl. Acad. Sci. U S A
101,
8180-8185 (2004).
Kipnis J, Nevo U, Panikashvili D, Alexandrovich A, Yoles E, Akselrod S,
Shohami E, Schwartz M. (2003) Therapeutic vaccination for closed head injury.
J.
Neurotrauma 20(6): 559-569 .
108

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Kipnis, J. and Schwartz, M. (2002) Dual Action of Glatiramer Acetate (Cop-
1) as a Treatment for Autoimmune Diseases and a Vaccine for Protective
Autoimmunity after CNS Injury. Trends Mol. Med. 8: 319-323.
Kipnis, J., Mizrahi, T., Yoles, E., Ben-Nun, A., Schwartz, M. & Ben-Nur, A.
J Neuroimmunol 130, 78-85 (2002a).
Kipnis, J., Mizrahi, T., Hauben, E., Shaked, I., Shevach, E. & Schwartz, M.
Neuroprotective autoimmunity: naturally occurring CD4+CD25+ regulatory T cells

suppress the ability to withstand injury to the central nervous system. Proc.
Natl.
Acad. Sci. USA 99, 15620-5 (2002b).
Kipnis J, Yoles E, Porat Z, Cohen A, Mor F, Sela M, Cohen IR, Schwartz M.
(2000), T cell immunity to copolymer 1 confers neuroprotection on the damaged
optic nerve: possible therapy for optic neuropathies, Proc Natl Acad Sci USA.
97:7446-7451.
Lafaille, J.J., Nagashima, K., Katsuki, M. & Tonegawa, S. High incidence of
spontaneous autoimmune encephalomyelitis in immunodeficient anti-myelin basic
protein T cell receptor transgenic mice. Cell 78, 399-408 (1994).
Ledeboer A, Breve JP, Poole S, Tilders FJ, Van Dam AM (2000)
Interleukin-10, interleukin-4, and transforming growth factor-beta
differentially
regulate lipopolysaccharide-induced production of pro-inflammatory cytokines
and
nitric oxide in co-cultures of rat astroglial and microglial cells. Glia
30:134-141.
Lepore, A. C. & Fischer, I. Exp Neurol194, 230-42 (2005).
Lessmann V, Gottmann K, Macicangio M, (2003) Neurotrophin secretion:
current facts and future prospect. Progress in Neurobiology 69:341-374.
Li, W.W., Setzu, A., Zhao, C., and Franklin, R.J. Minocycline-mediated
inhibition of microglia activation impairs oligodendrocyte progenitor cell
responses
and remyelination in a non-immiine model of demyelination. J Neuroimmunol
158:58-66 (2005).
Lu, P., Jones, L. L. & Tuszynski, M. H. Exp Neurol 191, 344-60 (2005).
Magavi SS, Leavitt BR, Macklls J (2000) I nduction of Neurogenesis in the
neocortex
of adult mice. Nature, 405:951-955.
109

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Magavi SS, Leavitt BR, Macklls J (2000) I nduction of Neurogenesis in the
neocortex of adult mice. Nature, 405:951-955.
Mason, J. L., Ye, P., Suzuki, K., D'Ercole, A. J. & Matsushima, G. K.
Insulin-like growth factor-1 inhibits mature oligodendrocyte apoptosis during
primary demyelination. J Neurosci 20, 5703-8 (2000).
McDonald, J. W., Becker, D., Holekamp, T. F., Howard, M., Liu, S., Lu, A.,
Lu, J., Platik, M. M., Qu, Y., Stewart, T. & Vadivelu, S. J Neurotrautna 21,
383-93
(2004).
Merrill, J. E., Ignarro, L. J., Sherman, M. P., Melinek, J. & Lane, T. E.
Microglial cell cytotoxicity of oligodendrocytes is mediated through nitric
oxide. J
Immunol 151, 2132-41 (1993).
Moalem, G., Leibowitz-Amit, R., Yoles, E., Mor, F., Cohen, IR. & Schwartz
M Autoimmune T cells protect neurons from secondary degeneration after central

nervous system axotomy. Nat. Med. 5, 49-55 (1999).
Moalem, G., Gdalyahu, A., Shani, Y., Otten, U., Lazarovici, P., Cohen, IR.
& Schwartz, M.. Production of neurotrophins by activated T cells: implications
for
neuroprotective autoimmunity. J Autoiminun 15, 331-45 (2000).
Monje, M. L., Mizumatsu, S., Fike, J. R. & Palmer, T. D. Irradiation induces
neural precursor-cell dysfunction. Nat Med 8, 955-62 (2002).
Monje, M. L., Toda, H. & Palmer, T. D. Inflammatory blockade restores
adult hippocampal neurogenesis. Science 302, 1760-5 (2003).
Morris, R. J Neurosci Methods 11, 47 (1984).
Morshead, C. M. et al. Neural stem cells in the adult mammalian forebrain: a
relatively quiescent subpopulation of subependymal cells. Neuron 13, 1071-82
(1994).
Nakatomi, H., Kuriu, T., Okabe, S., Yamamoto, S., Hatano, O., Kawahara,
N., Tamura, A., Kirino, T. & Nakafuku, M. Ce11110, 429-41 (2002).
Ness, J. K. & Wood, T. L. Insulin-like growth factor I, but not neurotrophin-
3, sustains Akt activation and provides long-term protection of immature
110

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
oligodendrocytes from glutamate-mediated apoptosis. Mot Cell Neurosci 20, 476-
88
(2002).
Neumann, H., Misgeld, T., Matsumuro, K., and Wekerle, H. Neurotrophins
inhibit major histocompatibility class II inducibility of microglia:
involvement of
the p75 neurotrophin receptor. Proc. Natl. Acad. Sci. USA 95:5779-5784 (1998).
Newman MP, Feron F, Mackay-Sim A (2000) Growth factor regulation of
neurogenesis in adult olfactory epithelium. Neuroscience 99:343-350.
O'Kusky, J. R., Ye, P. & D'Ercole, A. J. Insulin-like growth factor-I
promotes neurogenesis and synaptogenesis in the hippocampal dentate gyrus
during
postnatal development. J Neurosci 20, 8435-42 (2000).
O'Rourke NA, Sullivan DP, Kaznowsky CE, Jacobs AA, McConnell SK (1995)
Tangential migration of neurons in the developing cerebral cortex. Development

121:2165-2176.
Packer, M. A. et al. Nitric oxide negatively regulates mammalian adult
neurogenesis. Proc Natl Acad Sci U S A100, 9566-71 (2003).
Pencea V, Bingaman KD, Wiegand SJ, Luskin MB (2001) Infusion of brain-
derived neurotrophic factor into the lateral ventricle of the adult rat leads
to new
neurons in the parenchyma of the striatum, septum, thalamus, and hypothalamus.
J
Neurosci. 1:6706-6717.
Picard-Riera N, Decker L, Delarasse C, Goude K, Nait-Oumesmar B, Liblau
R, Pham-Dinh D, Evercooren (2002) Experimental autoimmune encephalomyelitis
mobilized neuronal progenitors from the subventricular zone to undergo
oligodendrogenesis in adult mice. Proc. Natl. Acad. Sci. USA. 99:13211-13216.
Picard-Riera N, Nait-Oumesmar B, Baron-Van Evercooren A (2004)
Endogenous adult neural stem cells: limits and potential to repair the injured
central
nervous system. J Neurosci. Res. 76:223-231.
Pluchino, S., Quattrini, A., Brambilla, E., Gritti, A., Salani, G., Dina, G.,
Galli, R., Del Carro, U., Amadio, S., Bergami, A., Furlan, R., Comi, G.,
Vescovi, A.
L. & Martino, G. Injection of adult neurospheres induces recovery in a chronic
model of multiple sclerosis. Nature 422, 688-94 (2003).
111

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Popovich, P. G., Wei, P. & Stokes, B. T. Cellular inflammatory response
after spinal cord injury in Sprague-Dawley and Lewis rats. J Comp Neurol 377,
443-64 (1997).
Popovich, P. G. et al. The neuropathological and behavioral consequences of
intraspinal microglial/macrophage activation. 1 NeuropathoL Exp. NeuroL 61,
623-
33 (2002).
Rapalino, O., Lazarov-Spiegler, O., Agranov, E., Velan, G. J., Yoles, E.,
Fraidakis, M., Solomon, A., Gepstein, R., Katz, A., Belkin, M., Hadani, M. &
Schwartz, M. Nat Med 4, 814-21 (1998).
Schori, H., Yoles, E. & Schwartz, M. T-cell-based immunity counteracts the
potential toxicity of glutamate in the central nervous system. 1 Neuroimmunol.
119,
199-204 (2001).
Schori H, Kipnis J, Yoles E, WoldeMussie E, Ruiz G, Wheeler LA,
Schwartz M. (200 lb) Vaccination for protection of retinal ganglion cells
against
death from glutamate cytotoxicity and ocular hypertension: implications for
glaucoma. Proc Natl Acad Sci U S A 98:3398-3403.
Schlegel PG, Aharoni R, Chen Y, Chen J, Teitelbaum D, Arnon R, Sela M,
Chao NJ. (1996) A synthetic random copolymer with promiscuous binding to class

II MHC molecules inhibits T-cell proliferative response to major and minor
histocompatibility antigens in vitro and confers the capacity to prevent
murine graft-
versus-host disease in vivo. Proc. Natl. Acad. Sci. USA. 93: 5061-6.
Schwartz, M. & Hauben, E. T cell-based therapeutic vaccination for spinal
cord injury. Prog Brain Res 137, 401-6 (2002).
Schwartz, M. & Kipnis, J. Harm or heal - divergent effects of autoimmune
neuroinflammation?. Response from Schwartz and Kipnis. Trends Immunol 24, 7-8
(2003).
Schwartz, M. & Kipnis, J. Autoimmunity on alert: naturally occurring
regulatory CD4(+)CD25(+) T cells as part of the evolutionary compromise
between
a 'need' and a 'risk'. Trends Immunol. 23, 530-4 (2002).
112

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Schwartz M, Kipnis J (2002) Multiple sclerosis as a by-product of the failure
to sustain protective autoimmunity: a paradigm shift. Neuroscientist 8: 405-
413.
Schwartz, M., Shaked, I., Fisher, J., Mizrahi, T. & Schori, H. Protective
autoimmunity against the enemy within: fighting glutamate toxicity. Trends
Neurosci. 26, 297-302 (2003).
Setoguchi, T., Nakashima, K., Takizawa, T., Yanagisawa, M., Ochiai, W.,
Okabe, M., Yone, K., Komiya, S. & Taga, T.Treatment of spinal cord injury by
transplantation of fetal neural precursor cells engineered to express BMP
inhibitor.
Exp Neurol 189, 33-44 (2004).
Shaked, I., Porat, Z., Gersner, R., Kipnis, J. & Schwartz, M. Early activation
of microglia as antigen-presenting cells correlates with T cell-mediated
protection
and repair of the injured central nervous system. J Neuroimmunol 146, 84-93
(2004).
Shaked, I., Tchoresh, D., Gersner, R., Meiri, G., Mordechai, S., Xiao, X.,
Hart, R. P. & Schwartz, M. J Neurochem 92, 997-1009 (2005).
Shors, T.J. et al. Neurogenesis in the adult is involved in the formation of
trace memories. Nature 410, 372-6 (2001).
Shors, T.J., Townsend, D.A., Zhao, M., Kozorovitskiy, Y. & Gould, E.
Neurogenesis may relate to some but not all types of hippocampal-dependent
learning. Hippocampus 12, 578-84 (2002).
Snyder, J.S., Hong, N.S., McDonald, R.J. & Wojtowicz, J.M. A role for adult
neurogenesis in spatial long-term memory. Neuroscience 130, 843-52 (2005).
Stollg G, Jander S (1999) The role of microglia and macrophages in the
pathophsiology of the CNS. Progress in Neurology 58:233-247
van Praag, H., G. Kempermann, F. H. Gage, Nat Rev Neurosci 1, 191 (2000).
van Praag, H., Christie, B.R., Sejnowski, T.J. & Gage, F.H. Running
enhances neurogenesis, learning, and long-term potentiation in mice. Proc Natl

Acad Sci USA 96, 13427-31 (1999).
113

CA 02588908 2007-05-29
WO 2006/057003 PCT/1L2005/001275
Yoles, E., Hauben, E., Palgi, O., Agranov, E., Gothilf, A., Cohen, A.,
Kuchroo, V., Cohen, I. R., Weiner, H. & Schwartz, M. Protective autoimmunity
is a
physiological response to CNS trauma. J Neurosci 21, 3740-8 (2001a).
Yoles, E., Friedmann, I., Barouch, R., Shani, Y. & Schwartz, M. J
Neurotrauma 18, 339-49 (2001b).
Zhao, X., Ueba, T., Christie, BR., Barkho, B., McConnell, MJ., Nakashima,
K., Lein, ES., Eadie, BD., Willhoite, AR., Muotri, AR., Summers, RG., Chun,
J.,
Lee, KF. & Gage, FH. Mice lacking methyl-CpG binding protein 1 have deficits
in
adult neurogenesis and hippocampal function. Proc Natl Acad Sci U S A100, 6777-

82 (2003).
Zielasek J, Tausch M, Toyka KV, Hartung HP. Production of nitrite by
neonatal rat microglial cells/brain macrophages. Cell Immunol 141:111-120
(1992).
114

DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPREND PLUS D'UN TOME.
CECI EST LE TOME 1 DE 2
NOTE: Pour les tomes additionels, veillez contacter le Bureau Canadien des
Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.
THIS IS VOLUME 1 OF 2
NOTE: For additional volumes please contact the Canadian Patent Office.

Representative Drawing

Sorry, the representative drawing for patent document number 2588908 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2014-09-23
(86) PCT Filing Date 2005-11-29
(87) PCT Publication Date 2006-06-01
(85) National Entry 2007-05-29
Examination Requested 2010-11-25
(45) Issued 2014-09-23

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $459.00 was received on 2021-11-15


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2022-11-29 $253.00
Next Payment if standard fee 2022-11-29 $624.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2007-05-29
Maintenance Fee - Application - New Act 2 2007-11-29 $100.00 2007-11-06
Registration of a document - section 124 $100.00 2008-05-23
Registration of a document - section 124 $100.00 2008-05-23
Registration of a document - section 124 $100.00 2008-05-23
Registration of a document - section 124 $100.00 2008-05-23
Registration of a document - section 124 $100.00 2008-05-23
Maintenance Fee - Application - New Act 3 2008-12-01 $100.00 2008-11-20
Maintenance Fee - Application - New Act 4 2009-11-30 $100.00 2009-11-04
Maintenance Fee - Application - New Act 5 2010-11-29 $200.00 2010-11-19
Request for Examination $800.00 2010-11-25
Maintenance Fee - Application - New Act 6 2011-11-29 $200.00 2011-11-29
Maintenance Fee - Application - New Act 7 2012-11-29 $200.00 2012-10-30
Maintenance Fee - Application - New Act 8 2013-11-29 $200.00 2013-10-23
Final Fee $756.00 2014-05-15
Maintenance Fee - Patent - New Act 9 2014-12-01 $200.00 2014-11-19
Maintenance Fee - Patent - New Act 10 2015-11-30 $250.00 2015-11-23
Maintenance Fee - Patent - New Act 11 2016-11-29 $450.00 2016-12-13
Maintenance Fee - Patent - New Act 12 2017-11-29 $250.00 2017-11-21
Maintenance Fee - Patent - New Act 13 2018-11-29 $250.00 2018-11-19
Maintenance Fee - Patent - New Act 14 2019-11-29 $250.00 2019-11-18
Maintenance Fee - Patent - New Act 15 2020-11-30 $450.00 2020-11-25
Maintenance Fee - Patent - New Act 16 2021-11-29 $459.00 2021-11-15
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
YEDA RESEARCH AND DEVELOPMENT CO. LTD
Past Owners on Record
AHARONI, RINA
ARNON, RUTH
BUTOVSKY, OLEG
EILAM, RAYA
EISENBACH-SCHWARTZ, MICHAL
KIPNIS, JONATHAN
RON, NOGA
ZIV, YANIV
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Cover Page 2007-08-20 2 37
Abstract 2007-05-29 1 67
Claims 2007-05-29 7 351
Drawings 2007-05-29 51 7,288
Description 2007-05-29 116 6,649
Description 2007-05-29 12 193
Claims 2007-05-30 6 293
Description 2010-12-07 116 6,571
Description 2010-12-07 12 195
Claims 2010-12-07 4 158
Claims 2012-11-02 1 24
Description 2012-11-02 116 6,541
Description 2012-11-02 12 195
Claims 2013-07-10 1 20
Cover Page 2014-08-26 2 39
Correspondence 2008-05-23 2 54
Assignment 2008-05-23 6 272
PCT 2007-05-29 3 91
Assignment 2007-05-29 4 101
Prosecution-Amendment 2007-05-29 8 329
Correspondence 2007-12-18 1 26
Fees 2007-11-06 1 34
Fees 2008-11-20 1 34
Fees 2009-11-04 1 34
Prosecution-Amendment 2010-11-25 1 37
Prosecution-Amendment 2010-12-07 15 657
Fees 2011-11-29 1 163
Correspondence 2014-08-05 1 34
Correspondence 2014-08-05 1 28
Prosecution-Amendment 2012-06-28 4 183
Prosecution-Amendment 2012-11-02 10 375
Prosecution-Amendment 2013-01-23 2 78
Prosecution-Amendment 2013-07-10 5 123
Fees 2013-10-23 1 33
Correspondence 2014-03-10 3 84
Correspondence 2014-05-05 4 188
Correspondence 2014-05-15 2 99
Correspondence 2014-05-29 1 23
Correspondence 2014-05-29 1 27
Correspondence 2014-07-24 1 25